Skip to main content

REVIEW article

Front. Bioeng. Biotechnol., 23 October 2019
Sec. Nanobiotechnology
Volume 7 - 2019 | https://doi.org/10.3389/fbioe.2019.00261

Hazard Assessment of Polymeric Nanobiomaterials for Drug Delivery: What Can We Learn From Literature So Far

  • 1Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
  • 2Laboratory for Technology and Society, Empa Swiss Laboratories for Materials Science and Technology, St. Gallen, Switzerland
  • 3School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
  • 4Laboratory for Particles-Biology Interactions, Empa Swiss Laboratories for Materials Science and Technology, St. Gallen, Switzerland
  • 5Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal

The physicochemical properties of nanobiomaterials, such as their small size and high surface area ratio, make them attractive, novel drug-carriers, with increased cellular interaction and increased permeation through several biological barriers. However, these same properties hinder any extrapolation of knowledge from the toxicity of their raw material. Though, as suggested by the Safe-by-Design (SbD) concept, the hazard assessment should be the starting point for the formulation development. This may enable us to select the most promising candidates of polymeric nanobiomaterials for safe drug-delivery in an early phase of innovation. Nowadays the majority of reports on polymeric nanomaterials are focused in optimizing the nanocarrier features, such as size, physical stability and drug loading efficacy, and in performing preliminary cytocompatibility testing and proving effectiveness of the drug loaded formulation, using the most diverse cell lines. Toxicological studies exploring the biological effects of the polymeric nanomaterials, particularly regarding immune system interaction are often disregarded. The objective of this review is to illustrate what is known about the biological effects of polymeric nanomaterials and to see if trends in toxicity and general links between physicochemical properties of nanobiomaterials and their effects may be derived. For that, data on chitosan, polylactic acid (PLA), polyhydroxyalkanoate (PHA), poly(lactic-co-glycolic acid) (PLGA) and policaprolactone (PCL) nanomaterials will be evaluated regarding acute and repeated dose toxicity, inflammation, oxidative stress, genotoxicity, toxicity on reproduction and hemocompatibility. We further intend to identify the analytical and biological tests described in the literature used to assess polymeric nanomaterials toxicity, to evaluate and interpret the available results and to expose the obstacles and challenges related to the nanomaterial testing. At the present time, considering all the information collected, the hazard assessment and thus also the SbD of polymeric nanomaterials is still dependent on a case-by-case evaluation. The identified obstacles prevent the identification of toxicity trends and the generation of an assertive toxicity database. In the future, in vitro and in vivo harmonized toxicity studies using unloaded polymeric nanomaterials, extensively characterized regarding their intrinsic and extrinsic properties should allow to generate such database. Such a database would enable us to apply the SbD approach more efficiently.

Introduction

Over the last decades, several nanomaterials (NMs) have been developed and studied as promisor drug delivery vehicles and medical devices, including magnetic, metallic, ceramic and polymeric nanomaterials. At present, there is fragile consensus regarding the “nano” definition among different regulatory organizations. In detail, considering medical regulatory authorities, such as the European Medicines Agency (EMA) or the United States Food and Drug Administration (FDA) some considerations can be made. In a reflection paper about nanotechnology-based medicinal products for human use published in 2006, EMA defined nanotechnology as “the production and application of structures, devices and systems by controlling the shape and size of materials at nanometer scale,” considering that “the nanometer scale ranges from the atomic level at around 0.2 nm (2 Å) up to around 100 nm” (European Medicines Agency, 2006). On its turn, FDA guidance for considering whether an FDA-regulated product involves the application of nanotechnology (Food Drug Aministration, 2014) refers that it should be considered “the evaluation of materials or end products engineered to exhibit properties or phenomena attributable to dimensions up to 1,000 nm, as a means to screen materials for further examination and to determine whether these materials exhibit properties or phenomena attributable to their dimension(s) and associated with the application of nanotechnology.” Therefore, for the context of academic research and to the context of this review the following definition of nanomaterial applies: materials in the size range of 1 nm to 1,000 nm and a function or mode of action based on its nanotechnological properties. In addition, by “nanobiomaterial” we considered NMs intended to interact with biological systems. The application of nanobiomaterials in the medicine field present several advantages as they can (Moritz and Geszke-Moritz, 2015; Banik et al., 2016):

• Transport higher drug payloads

• Enable targeted drug delivery

• Increase the bioavailability of poorly water-soluble drugs

• Promote controlled drug delivery

• Increase the stability of drugs in biological fluids

• Increase drug circulation time in the body

• Confer drugs protection from biological fluids

• Permeate through various biological barriers

• Enable surface modifications to increase interaction with biological targets.

Considering polymeric NMs in particular, they can be assembled in different pharmaceutical nanosystems, such as nanoparticles (NPs), dendrimers, polymeric micelles and drug conjugates (Bhatia, 2016). On its turn, polymeric NPs comprise both vesicular systems (nanocapsules) and matrix systems (nanospheres) (Bhatia, 2016). The polymeric nature of these NMs provides additional advantages that are worth exploring, such as enhanced biocompatibility, biodegradability and low immunogenicity (Egusquiaguirre et al., 2016; Rana and Sharma, 2019).

All considered, most of these advantages are frequently attributed to their distinctive size which contributes to their high surface area to mass ratio, and is also responsible for the different toxicokinetic fate of the NMs (Landsiedel et al., 2012; Boyes et al., 2017). Indeed, small sizes facilitate cell uptake, penetration through endothelial and epithelial cells, interaction with tissues and accumulation in the liver, kidney and spleen (Khan and Shanker, 2015). The increased cellular interaction can have a modulatory effect on the immune system, triggering inflammation, increased susceptibility to infectious diseases, or even to autoimmune diseases or cancer (Kononenko et al., 2015).

The unique physicochemical properties of the NMs restricts the extrapolation of toxicological data from raw materials, and makes it necessary to have specific toxicological studies adequate to the nanoscale (Ge et al., 2011). Moreover, there is a need for specific and optimized methods for NMs toxicity evaluation, since interactions between NMs and current toxicity testing protocols can lead to false positive or false negative results (Khan and Shanker, 2015; Kononenko et al., 2015).

Understanding the toxicokinetics of NMs and their modulation of the immunological system is necessary to implement their Safe-by-Design based on the literature. This is an up-to-date subject, currently widely discussed among the scientific community, but most commonly for metallic NM (Gatto and Bardi, 2018; Kanwal et al., 2019).

Therefore, the objective of this review is to summarize what is known about the toxic effects of polymeric NMs, with special focus on polymeric NPs that could be correlated to human health risks. We intend to identify the analytical and biological tests described in the literature used to assess NMs toxicity and to evaluate and interpret the available results. Furthermore, we intend to understand the obstacles and challenges related to the nanomaterial testing that are still preventing a harmonized regulation on polymeric NMs for drug delivery and biomedical applications.

We started this review by discussing the pillars of human health risk assessment: exposure assessment and hazard assessment. Next, in order to analyze the state of the art about the toxic effects of polymeric NMs, peer reviewed original research articles from the last 10 years were analyzed and discussed, addressing the following endpoints: (1) in vivo toxicity (acute and repeated-dose), (2) oxidative stress, (3) inflammation, (4) genotoxicity, (5) toxicity on reproduction and (6) hemolysis. Importantly, articles were carefully examined regarding minimal characterization parameters, such as chemical composition, particle size, surface charge and endotoxin contamination (when relevant).

Pillars for Human Health Risk Assessment

To perform human health risk assessment of any material is necessary to integrate the exposure assessment with hazard assessment. The first intends to determine routes of exposure and estimate exposure dosages (dose, duration and frequency) while the second intends to characterize the possible hazards (toxic effects) of polymeric NMs when in contact with the human body.

Exposure Assessment

Human exposure to polymeric NMs should be considered in the context of intentional nanomedicine applications, and in the context of occupational exposures of workers during the manufacturing processes, testing methods, distribution and handling/administration of polymeric NMs. Moreover, it cannot be disregarded situations where misuse and overuse are easily attained (Sayes et al., 2016). While in nanomedicine exposure scenarios, the administration route, the dose and duration of the exposure are well-defined, occupational exposure can happen through multiple and non-expected routes (Figure 1) and result in potentially cumulative levels of exposure and organ accumulation, whose impact in human health might be very different from the one predicted (Sayes et al., 2016). In fact, working with NMs involves challenges different from when working with bulk size materials, since they have increased ability to enter the human body, particularly through the respiratory airways, and to be translocated to the bloodstream and different organs (Yah et al., 2012). The lack of testing methods to detect and quantify the unintentional absorbed cumulative doses of these materials in the organism is currently, one of the main difficulties for designing predictive toxicological assays for occupational exposures. Therefore, exposure modeling arises as one alternative to allow occupational risk assessment. In the context of the FP7 NanoReg project a number of risk assessment tools for manufactured NMs, such as the CB NanoTool, the Nanosafer, and the Stoffenmanager-Nano have been examined and a new two-box nano specific exposure model (I-Nano) has been implemented (Jiménez et al., 2016). However, the need to rely on detailed input data (rate of particulate release from the source as well as the particle size distribution) which is not always available and its only application to inhalable exposures are some of the limitations present (Jiménez et al., 2016).

FIGURE 1
www.frontiersin.org

Figure 1. Hypothetical scenarios of exposure: comparison between the administration route and doses foreseen in medical applications and the exposure routes and cumulative doses difficult to predict in unintentional exposures, such as occupational scenarios.

In the main, the NM routes of administration and exposure include respiratory, oral, ocular, dermal, and parenteral (injectable and implantable), each route presenting its own biodistribution pattern, resulting in different effects on human health. Indeed, the same composition, size and surface charge of the polymeric NM, might produce a different effect only by changing the exposure route (Sharma et al., 2016; Boyes et al., 2017). Importantly, it cannot be disregarded that the characteristics of the individual exposed, such as its age and health status, might also influence the NMs effect (Boyes et al., 2017). Table 1 below summarizes the most common administration/exposure routes and the most important characteristics of NMs related to each one.

TABLE 1
www.frontiersin.org

Table 1. Common routes of administration/exposure: important considerations relating nanomaterials characteristics and the various routes of exposure (Agrawal et al., 2014; Blanco et al., 2015; Date et al., 2016; Palmer and DeLouise, 2016; Boyes et al., 2017).

Hazard Assessment

The NMs toxic effects might occur in the administration site or they can result from the nano-sized materials ability to cross biological barriers (mucosal barriers, air-blood barrier, blood-brain barrier, placenta barrier) reaching cells and tissues that are generally protected from bulk size materials (Buzea et al., 2007; Ai et al., 2011). This improved penetration of nanoparticles may increase the toxicity, but at the same time be advantageous in order to improve current therapies.

The uncertainties about using NMs for drug delivery and other biomedical applications result mainly from particle size reduction which is linked to increased reactivity and augmented toxicity (Ai et al., 2011). Nonetheless, several other properties can contribute to the effects of these nano-sized delivery systems, such as chemical composition, hydrophobicity/hydrophilicity, surface charge or shape. In the literature, there is a significant amount of data relating physicochemical features of NMs with cellular interaction, biodistribution, cytotoxicity and immune system activation, as reviewed elsewhere (Fröhlich, 2012; Ma et al., 2013; Salatin et al., 2015; Hoshyar et al., 2016; Jindal, 2017; Zhang et al., 2017). Nevertheless, general conclusions indicating toxicity trends for a specific nanoparticle physicochemical property, are limited to cautious hypotheses, only verified in particular scenarios (i.e., depending on the administration route, dose metrics, etc.). A review published in 2014 by Gatoo et al. (2014) discusses the correlation between the physicochemical properties of NMs and its toxicity. Briefly, smaller particles are often correlated with a higher toxicity, due to their increasing ability to cross biological barriers and reach different organs without being recognized by the reticuloendothelial system (RES) (Gatoo et al., 2014). Other characteristics, such as the non-spherical shape or the positive surface charge are also believed to contribute to an increased toxicity of NMs (Gatoo et al., 2014). Importantly, most of these conclusions are based on studies using inorganic NMs. Since chemical composition is one of the variables affecting the NMs toxicity, different behaviors can derive from the polymer composition and therefore, extensive extrapolations among all classes of NMs should be avoided. Moreover, most toxicity trends consider one characteristic at a time, but it is important to consider a holistic approach of the NM: all physicochemical characteristics are interconnected and together will influence its toxicological profile.

The key aspect to test polymeric NM for human toxic effects is the simulation of realistic human exposures. Those scenarios are difficult to simulate mainly due to: (1) the difficulty on transposing accurately human effective doses to in vitro settings; and (2) the difficulty to have complex in vitro systems, based on human cells or primary cell lines, that mimic the physiological complexity of the human body and its interaction with the materials (Sharma et al., 2016). Actually, most of the results of the application of in vitro studies to polymeric NMs might not reflect the realistic exposures, since the tests are performed at much higher concentrations than those that can be achieved in in vivo experiments (Landsiedel et al., 2017). Moreover, in vitro testing commonly use mass-based exposure metrics, which is believed to be a limiting factor, as particle number, surface areas and the formed agglomerates in suspension greatly influence the effective concentration delivered to cells (Hinderliter et al., 2010; DeLoid et al., 2014).

The intrinsic and distinctive characteristics inherent to the nanoscale dimension, might interfere with reagents and detection methods of in vitro assays recommended for bulk materials (Dobrovolskaia et al., 2009). For instance, NMs may bind to the marker enzyme lactate dehydrogenase (LDH) or they may interact with dyes and dye products, such as neutral red and the tetrazolium salt (MTT) (Landsiedel et al., 2017). On the other hand, polymeric NMs also go through modifications when in contact with biological matrices, such as: bio-corona formation, aggregation/agglomeration, dissolution, generation of new nano-sized particles (as a result of ionic salvation or degradation of surface coatings) (Sharma et al., 2016). These transformations of the NM can interfere with its toxicological effect, and most of the times are not considered during in vitro testing. Lastly, the selection of relevant positive and negative nano-sized controls is most of the times ignored, mainly because there is no clear knowledge-base on the toxicity (and especially immunotoxicity) of the different NMs (Dobrovolskaia and McNeil, 2013).

It is widely accepted that in vitro assays based on cell lines are an inexpensive and direct method to evaluate nanoparticle related toxicity in target tissues. However, results significantly depend on the chosen cell line (commonly immortalized cancer cells), incubation time, cell culture media or cell culture supplementation (Lorscheidt and Lamprecht, 2016). For instance, cell culture media supplementation with serum is highly likely to induce a protein corona in the surface of positively charged nanoparticles, changing its size and zeta potential, and therefore modifying the nanoparticle-cell interaction and uptake, and ultimately its biological effect (Khang et al., 2014; Lorscheidt and Lamprecht, 2016).

Overall, despite the great effort in developing high-throughput in vitro assays, there is still much variables to accurately mimic real exposure scenarios, and the results are often in disagreement with those of animal studies (DeLoid et al., 2014). Even so, nanotechnology laboratories are still searching for the best in vitro assays to replace in vivo testing and predict real exposure scenarios. This issue has been extensively discussed by Dobrovolskaia and McNeil (2013).

The urge to replace in vivo testing of toxicity, is motivated by the high costs and relatively low throughput of the assays, the inter-species variability particularly on the structure and function of the immune system, the low sensitivity of standard in vivo toxicity tests toward mild immunomodulation reactions, and most importantly, the ethical concerns about animal use (Dobrovolskaia and McNeil, 2013).

Altogether, it is widely accepted that efficient and cost-effective toxicological testing is required (DeLoid et al., 2014). For that reason, international organizations including OECD and ISO have developed official papers with considering the NMs properties and their influence on testing methods (Sharma et al., 2016; Dusinska et al., 2017).

In 2006, the OECD started a nanosafety programme overseen a Working Party on Manufactured Nanomaterials (WPMN), which aims to promote international cooperation on the human health and environmental safety of manufactured NMs, and involves the safety testing and risk assessment of manufactured NMs. Over the years they have published numerous reports and some test guidelines which are published in the OECD Series on the Safety of Manufactured Nanomaterials to provide up-to-date information on the OECD activities in this area (OECD1).

In 2005, the Technical Committee ISO/TC 229 was created. It aims at the standardization in the field of nanotechnologies. The specific tasks of this committee include developing standards for terminology and nomenclature, metrology and instrumentation, test methodologies, modeling and simulations, and science-based health, safety, and environmental practices (Behzadi et al., 2014). Over the years, the committee has published several standards, from which we can highlight the recent ISO/TS 19006:2016 [Nanotechnologies-5-(and 6)-Chloromethyl-2′,7′-Dichloro-dihydrofluorescein diacetate (CM-H2DCF-DA) assay for evaluating nanoparticle-induced intracellular reactive oxygen species (ROS) production in RAW 264.7 macrophage cell line] and the ISO 19007:2018 (Nanotechnologies–in vitro MTS assay for measuring the cytotoxic effect of nanoparticles), discussed below (Bazile et al., 1995; Behzadi et al., 2017). In addition to the specific standards generated by this committee, in 2017, the part 22—Guidance on nanomaterials, was implemented in ISO 10993 (Biological evaluation of medical devices) (Barratt, 2000). Although this technical report represents the current technical knowledge related to NMs for medical devices it does not contain detailed testing protocols.

An important contribution to this field is being given by the US National Cancer Institute Nanotechnology Characterization Laboratory, whose main objective is to facilitate the development and translation of nanoscale particles and devices for clinical applications. In fact, they have described several protocols for in vitro characterization as well as for in vivo, and for the physicochemical characterization of NMs (Assay Cascade Protocols—https://ncl.cancer.gov/resources/assay-cascade-protocols). In parallel, the European Nanomedicine Characterization Laboratory (EUNCL) is also developing standard operating procedures (SOPs) to allow the physical, chemical, in vitro and in vivo testing of nanobiomaterials (http://www.euncl.eu/).

Hazard Characterization of Polymeric Nanomaterials—Literature Review

NMs toxicity should be evaluated by in vivo and in vitro assays considering its effect in the host physiological and immunological integrity (Yildirimer et al., 2011). Most of in vitro assays available for testing a NM toxicological effects are focused on the molecular mechanisms underlying toxicity (i.e., oxidative stress generation and inflammation), while in vivo assays, particularly acute and repeated dose toxicity assays assess the effects on vital organ functions [i.e., biomarkers of liver function, such as aspartate aminotransferase (AST) and alanine aminotransferase (ALT)].

Table 2 summarizes the studies collected from the literature of the last 10 years, assessing the toxicity of polymeric NMs for the endpoints studied. The polymers considered for analysis were chitosan, polylactic acid (PLA), polyhydroxyalkanoate (PHA), poly(lactic-co-glycolic acid) (PLGA) and policaprolactone (PCL). From the table systematization we can highlight three main issues: (1) chitosan based NPs are the most studied polymeric NMs followed by PLGA based NPs; (2) the different colors illustrating the generation or absence of effect for each endpoint according to the different studies, reflects the inconsistency in the results found for the same type of NM; (3) No data on PHA based NMs is available regarding those endpoints. The inconsistent results must be carefully analyzed because in fact they may be complementary results, as the NM characteristics, their concentrations, the cellular and animal models used and even the experimental methodology are significantly different among authors. Therefore, in the next sub-chapters each endpoint and respective studies will be discussed in detail in an attempt to scrutiny possible toxicity trends for polymeric NMs. To note, over the following discussion, the effect of some other polymers, such as alginate, polyethylene glycol (PEG), pluronic and polyvinyl alcohol (PVA) are addressed as they are often used as surface coatings and blends in chitosan, PLGA, PLA and PCL based nanomaterials.

TABLE 2
www.frontiersin.org

Table 2. Systematization of the toxicity results described in the literature for chitosan, PLA, PHA, PLGA, and PCL nanomaterials.

In vivo Toxicity Studies

To study the toxicity of the NMs and to identify possible risks to the human health, researchers perform in vivo tests in animals (most time non-primates) to evaluate acute and repeated-dose (subacute, sub-chronic or chronic) toxicity. These studies, although highly valuable to understand the adsorption, distribution, metabolism and excretion (ADME) of the NMs as well as the immune system interactions, should be limited to a minimum according to the 3Rs strategy (replacement, reduction and refinement) (Oostingh et al., 2011; Dusinska et al., 2017). To note, in 2018, OECD guidelines for the testing of chemicals were adapted to accommodate the testing of NMs (OECD, 2018b,c).

As illustrated in Table 3, the available research articles testing in vivo the toxicity of NMs are characterized by a great variability between the rodent's species (or other animals, such as carps) used in the assays, the number of days (for the repeated-dose toxicity studies) and even for the endpoints that are analyzed. Some of the most reported endpoints are the clinical appearance of the animal, clinical signs of infection, hematological parameters, serum hemoglobin levels and albumin/globulin ratio, organ weights, and enhanced histopathology evaluation different organs (Dusinska et al., 2017).

TABLE 3
www.frontiersin.org

Table 3. Review of original articles assessing in vivo the toxicity of polymeric nanoparticles.

As already stated, chitosan NMs are the most studied polymeric NMs regarding toxicity. Several studies were found in the literature evaluating the toxicity of blend chitosan NPs upon repeated oral administrations. Despite the great heterogeneity among the used NPs (chitosan/alginate NPs, chitosan/glutamic acid NPs, oleoyl-carboxy methyl chitosan NPs, chitosan coated PLGA NPs and α-tocopherol succinate-g-carboxymethyl chitosan NPs), the animal models (Wistar and Sprague Dawley rats, ICR mice and Carps) and the dosing schedules (7–19 days), all revealed no in vivo toxicity (Sonaje et al., 2009; Liu et al., 2013; Jena and Sangamwar, 2016; Aluani et al., 2017; Maity et al., 2017; Radwan et al., 2017b; Sharma et al., 2017). Moreover, the conclusion of no toxicity was based on different evaluated parameters for each study, except for the histopathological analysis, which was performed in all studies (generally liver and intestine histopathology with no signs of tissue damage). Among these studies, only Sonaje et al. (2009), Maity et al. (2017), and Radwan et al. (2017b) have evaluated biochemical parameters in blood, and in common have tested serum alanine transaminase (ALT), alkaline phosphatase (ALP) and aspartate transaminase (AST) activities, and their results were in agreement (no changes in comparison to the control group). Moreover, chitosan based NPs lack of oral toxicity was also reported for single dose administrations (Mukhopadhyay et al., 2015; Leng et al., 2018). Therefore, considering these reports, we may hypothesize that chitosan NPs (as well as bulk chitosan Chang et al., 2014) do not present oral toxicity. On the other hand, although only 2 reports were found testing chitosan NPs toxicity through the injectable route (Yuan et al., 2015; Shan et al., 2017), a dose dependent toxicity was found, even though chitosan and chitosan NPs appear to be hemocompatible in some hemolysis assays (Fernandes et al., 2010; Lü et al., 2011; Wang et al., 2014; Kumar et al., 2017; Leng et al., 2018).

On its turn, PLGA NPs also exhibited no toxicity on repeated oral administration studies (Moraes Moreira Carraro et al., 2017; Sharma et al., 2017), as well as on the majority of intravenous (i.v.) administration studies (VasanthaKumar et al., 2014; Fasehee et al., 2016; Radwan et al., 2017a). Only one article described some toxicity when using danorubicin loaded PEG-PLL-PLGA NPs (Guo et al., 2015). Unfortunately, the formulations in those reports were loaded with the active drug and no information was given on blank NPs. Therefore, not only the effects might be associated with the drugs (rather than the NPs polymers or characteristics), but also no comparison on the dose of the NPs administered can be made between articles, as they only refer to the equivalent amount of drug administered. Similarly (Li et al., 2014), tested two mPEG-PLA NPs (with different copolymerization degrees) loaded with paclitaxel in beagle dogs by i.v. administration in the foreleg. Despite the results had revealed differences between the NPs, being the ones with the 50/50 ratio mPEG:PLA more toxic than the ones with the 40/60, no experiments were made with unloaded NPs, restricting the extrapolation of data.

Oxidative Stress

Reactive oxygen species (ROS) are produced during cellular metabolism in the forms of hydrogen peroxide (H2O2), superoxide anion (O2−•) and hydroxyl (OH) radicals (Ngo and Kim, 2014; Lorscheidt and Lamprecht, 2016). Besides its role in cell signaling and regulation, excessive oxidative stress can induce oxidative damage to cells through lipid peroxidation, DNA disruption, interference with signaling functions, gene transcription modulation and inadvertent enzyme activation, causing several health disorders, such as hypertensive, cardiovascular, inflammatory, aging, diabetes mellitus, and neurodegenerative and cancer diseases (Sharifi et al., 2012; Ngo and Kim, 2014; Lorscheidt and Lamprecht, 2016).

The most used probe to access ROS is the H2O2 specific 2′,7′-dichlorodihydrofluorescein diacetate (H2DCFDA or DCFH-DA), which diffuses freely through the cell membrane and is hydrolyzed inside the cells into H2DCF carboxylate anion form, which is in its turn non-permeable (Kalyanaraman et al., 2012; Oparka et al., 2016). Then, H2DCF is oxidized and results in the formation of the fluorescent product (DCF), which is excited at 495 nm and emits at 520 nm (Kalyanaraman et al., 2012; Oparka et al., 2016). Using this probe, the intracellular signal can be monitored by several techniques, such as confocal microscopy and flow cytometry (Kalyanaraman et al., 2012). During the H2DCF oxidation, there is a formation of a superoxide radical that can stimulate the auto-amplification of the DCF signal (Oparka et al., 2016). On the other hand, DCF is cell permeable, which means it leaks out of cells over time and can induce measurement errors depending on the analysis time (Lorscheidt and Lamprecht, 2016). A variant of the DCFH-DA probe is the 5-(and 6)-chloromethyl-derivative, that leads to the formation of fluorescent CM-DCF, which displays a lower passive leakage from the cell (Oparka et al., 2016). Alternatively, the fluorescence read-out can also be performed using a fluorescence microplate reader and in this situations errors can result from nanoparticle quenching effect over the DCF fluorescence (Aranda et al., 2013).

Free radical production is the highest in macrophages (Singh and Ramarao, 2013) which is in line with the protocol suggested in ISO/TS 19006:2016-Nanotechnologies-5-(and 6)-Chloromethyl-2′,7′-Dichloro-dihydrofluorescein diacetate (CM-H2DCF-DA) assay for evaluating nanoparticle-induced intracellular reactive oxygen species (ROS) production in RAW 264.7 macrophage cell line. Nonetheless, according to this ISO, other cell lines similar to RAW 264.7 (BEAS-2B, RLE-6TN, HEPA-1, HMEC and A10) can be used with due validations. In this technical specification, the protocol was validated for conducting the assay in 24 well-plates, for 6 and 24 h incubation with the NPs and controls, and 30 min incubation with the probe before flow cytometry analysis. To note, the recommendation is the use of Sin-1 as positive control (maximum ROS production due to cell death) and polystyrene NPs as negative control.

As it is possible to observe from Table 4, most studies reported in the literature do not use RAW 264.7 cells, neither do they employ 6 and 24 h incubation.

TABLE 4
www.frontiersin.org

Table 4. Review of original articles assessing oxidative stress induction by polymeric nanoparticles.

In detail, Grabowski et al. found a transient production of ROS with chitosan stabilized PLGA NPs in THP-1 cells (Grabowski et al., 2015), Sharma et al. verified an increased oxidative effect of oleanolic acid when delivered by chitosan coated PLGA NPs in MDAMB-231 cells (Sharma et al., 2017), Sarangapani et al. found an increase in ROS production in BCL2(AAA) Jurkat cells with chitosan NPs (Sarangapani et al., 2018) and Gao et al. found an increase in ROS production in zebrafish embryos incubated with chitosan NPs (Hu et al., 2011). In contrast, Bor et al. found a reduction in ROS production with plasmid loaded chitosan NPs and chitosan NPs in Hela, THP-1 and MDAMB-231 cells (Bor et al., 2016). These inconsistent results, obtained with different chitosan based nanomaterials, different cellular models and concentrations do not allow for a straightforward interpretation of the oxidative effect of nanoscale chitosan. Among these articles, only Sarangapani et al. compared the activity of chitosan NPs with bulk chitosan (at the same concentrations) and verified a similar but lower concentration dependent effect for the polymer (Sarangapani et al., 2018). Also, it is important to note, that the tested concentrations (10–50 μg/mL), caused increasing cell death as verified by the MTT assay, and therefore, the oxidative stress was the mechanism identified as responsible for cellular toxicity. In contrast, Bor et al. verified that chitosan NPs reduced ROS production in several cell lines (also tumor derived cells), but they used a concentration that did not cause cell death (Bor et al., 2016). Therefore, although at first sight the results are conflicting, they cannot be directly compared, but we can hypothesize that chitosan NPs might influence ROS production in a concentration dependent manner. One of the widely reported characteristics of bulk chitosan is its anti-oxidant activity, attributed to its scavenging activity against several radicals, such as hydroxyl (OH), superoxide anion (O2-), 1,1-diphenyl-2-picryl-hydrazy (DPPH) and alkyl (Ngo and Kim, 2014). This scavenging activity, has been widely demonstrated by cell-free in vitro assays (Je et al., 2004; Yen et al., 2008; Ngo and Kim, 2014). In fact, in the article discussed before (Sarangapani et al., 2018), although reporting that chitosan and chitosan NPs increased ROS production in BCL2(AAA) Jurkat cells, they also verified that the same concentrations increased free radical scavenging activity using chemical assays. Therefore, some compounds may demonstrate chemically some antioxidant activity, which is not verified at cellular and physiological level (Lü et al., 2010).

Regarding bare PLGA NPs its effect on ROS production was documented by 3 authors Platel, Singh, and Granbowski (Singh and Ramarao, 2013; Grabowski et al., 2015; Platel et al., 2016) all using different cellular models. Nevertheless, Platel tested only one low concentration of PLGA NPs (40 μg/mL) and found no effect on ROS production (Platel et al., 2016), while the other 2 authors found an increase in ROS production that was dose dependent (Singh and Ramarao, 2013; Grabowski et al., 2015). Curiously, both tested 1 mg/mL, but Singh et al. reported that this concentration quenched the fluorescence of the probe, therefore interfering with the results (Singh and Ramarao, 2013). On its turn, Grabowski et al. found that at the concentration of 1 mg/mL only a transient production of ROS was verified at 5 min after the incubation with PLGA NPs, and at longer incubation times, no significant ROS increase was verified (Grabowski et al., 2015). Although the authors do not explore this achievement, we could hypothesize that a similar interference as reported by Singh and Ramarao might be occurring.

Overall, not only PLGA NPs, but in general the polyester NPs appear to induce ROS production in a concentration dependent manner. Other studies confirm this effect for concentrations above 300 μg/mL (Singh and Ramarao, 2013; Legaz et al., 2016; Da Silva et al., 2019). Nevertheless, this conclusion has reservations since for instance, Da Silva et al. tested two different PLA NPs, and only one of these induced ROS production.

Inflammation

Presently, inflammation is acknowledged as a mechanism of immune defense and repair, in addition to its widely accepted role in passive cell injury and cell death (Wallach et al., 2013; Khanna et al., 2015). Interestingly, several molecules are associated with inflammation and cell death. For instance TNF-α, IL-1β, IL-6, IFN-γ, IL-17, IL-8, IL-2, GM-CSF, TGF-β, and IL-12 are examples of pro-inflammatory mediators frequently evaluated in the context of cellular toxicity induced by nanomaterials (Khanna et al., 2015; Lorscheidt and Lamprecht, 2016).

Regarding the methodologies, the enzyme-linked immunosorbent assay (ELISA) is widely applied as a simple mean to perform a qualitative and quantitative analysis of cytokines, chemokines, growth factors and immunoglobulins, with a spectrophotometric readout (Lorscheidt and Lamprecht, 2016). In this assay, the pro- and anti-inflammatory mediators are released into cell supernatant, which is collected and then analyzed. Therefore, the release of cytokines or other molecules by cells during the incubation with nanoparticles can be underestimated due to the nanoparticles ability to adsorb biomolecules at its surface (Lorscheidt and Lamprecht, 2016). Kroll et al. (2012) tested the potential interference of 4 types of engineered nanoparticles on IL-8 secretion, and verified that a specific pre-dispersion of TiO2 nanoparticles was able to reduce the measurable levels of the cytokine, under the assay conditions. Similarly, Guadagnini et al. (2013a), tested 4 types of nanoparticles in acellular conditions and verified that TiO2, SiO2, and Fe3O4 NPs decreased the cytokines levels due to surface adsorption. In the same experiment, PLGA-PEO NPs induced an apparent increase in GM-CSF levels, which the authors believe may be due to the stabilization of the peptides, their protection from proteolysis or by avoiding the interaction of this cytokine with the plastic of the culture plates (Guadagnini et al., 2013a). Although most of the reported interferences are for inorganic nanoparticles, these are good examples that can be overlooked when performing ELISA in cell supernatants previously incubated with polymeric nanoparticles. When studying pro- and anti-inflammatory molecules release due to NPs stimulation, it can be useful to previously study the adsorption or interaction of the NPs with the molecules (i.e., cytokine standards) in acellular conditions.

Alternatively, instead of measuring cell secreted pro- and anti-inflammatory molecules by ELISA, the mRNA levels inside the cell can be measured with RT-qPCR (Real-Time quantitative Polymerase Chain Reaction) or the intracellular levels of the cytokines can be measured by flow cytometry analysis using specific antibodies fluorescently labeled (Lorscheidt and Lamprecht, 2016). In the first alternative, however, an increase of mRNA expression does not necessarily lead to an increase of protein secretion (Guadagnini et al., 2013a).

Lastly, besides the masking/enhancing effect of NPs, the presence of contaminants, such as endotoxins can induce itself increased levels of pro-inflammatory molecules in cells (Oostingh et al., 2011). Endotoxins, commonly referred to as lipopolysaccharide (LPS), are present in the outer cell membrane of Gram negative bacteria and are released during multiple processes, such as cell death, growth and division (Magalhaes et al., 2007; Lieder et al., 2013). Therefore, due to the bacteria ability to growth and adapt in several environments, LPS is easily found in numerous media, including poor nutrient media (water, saline and buffers) and its removal is a struggle since it is highly resistant to extreme temperatures pHs (Magalhaes et al., 2007). LPS is comprised by a O-antigen region, a hydrophilic core oligosaccharide and a hydrophobic Lipid A (LipA) (Davydova et al., 2000; Magalhaes et al., 2007; Steimle et al., 2016). The lipid A structure, highly conserved, differs among bacterial species, and determines the molecule immunogenicity (Steimle et al., 2016). On the whole, LPS is a pathogen associated molecular pattern (PAMP), which is recognized and activates the mammalian innate immune system, leading for instance to cellular release of pro-inflammatory cytokines and free radicals, particularly by monocytes and macrophages (Yermak et al., 2006; Lieder et al., 2013; Steimle et al., 2016). Consequently, in vitro testing of LPS contaminated polymeric NMs might generate misleading results and false assumptions of bioactivity or toxicity, ultimately affecting the evaluation of possible human health effects (Lieder et al., 2013).

Table 5 summarizes the results found in the literature for polymeric NPs stimulation of cytokines.

TABLE 5
www.frontiersin.org

Table 5. Review of original articles assessing inflammatory cytokines induced by polymeric nanoparticles in different cells.

For chitosan NPs, it is interesting to notice that one author referred chitosan NPs induced several cytokines in BMDCs (Koppolu and Zaharoff, 2013), while other did not (Han et al., 2016). Nevertheless, in both papers, no endotoxin contamination was assessed, no concentrations of NPs were given and the chitosan polymers and NPs characteristics were not the same. Furthermore, it must be considered that cytokine secretion highly depends on the cellular model under study. Indeed, Koppolu and Zaharoff, upon stimulation with chitosan NPs, reported the production of IL-1β in BMDCs and the absence of the same cytokine in RAW 264.7 (Koppolu and Zaharoff, 2013).

The fact that no endotoxin control was made in both papers can rise several questions, mainly in the results that suggest a positive stimulation of chitosan NPs. Chitosan has a cationic charge, resultant from the N-acetyl group removal during chitin deacetylation. This positive charge, mediates for instance the electrostatic interactions with cargo molecules, allowing high loading efficacies, but it also enables chitosan interactions with the negatively charged phosphate, pyrophosphate, and carboxylic groups of LPS (Davydova et al., 2000). Actually, chitosan has been used as a selective filtration membrane for endotoxin removal due to these extensive interactions (Machado et al., 2006; Lieder et al., 2013).

But not only chitosan should be evaluated regarding endotoxin contamination. For instance, Grabowski et al. have published two reports, comparing the inflammatory ability of different PLGA NPs based on the in vitro assessment of cytokines, such as IL-6, TNF-α, IL-8 and MCP-1 (Grabowski et al., 2015, 2016). The differences among PLGA NPs resulted from the inclusion of chitosan, PVA and P68 in order to obtain, positive, neutral and negatively charged particles. In one of the reports the authors do not evaluate or discuss the presence of endotoxin contamination in the formulations (Grabowski et al., 2015). Nonetheless, in the other report, using the same methods and polymers, the authors mentioned that all formulations presented 0.1 to 0.3 EU/mL of LPS depending on the concentration used (Grabowski et al., 2016). In both reports, this information was imperative, since the authors tested IL-8, IL-6 and TNF-α, cytokines whose production is induced by LPS (Agarwal et al., 1995; Grabowski et al., 2016). Therefore, despite their conclusions, as illustrated in Table 5 (Grabowski et al., 2015, 2016), and despite the authors attribute the observed effects to the nanoparticulate form of the formulations, the effect of LPS contamination might be interfering with the results. A simple control that could be adopted in this situation, was to use the LPS concentration the authors quantified in the formulations, incubate with the cell and assess the cytokine secretion. In these articles, the relationship between the 0.1–0.3 EU/ml of contamination and the 0.1–10 μg/mL of LPS as control was not given, and therefore, no further conclusions could be drawn regarding the effect of the LPS contamination in the formulations. Another relevant aspect to highlight, is the fact that nanoparticles, particularly polymeric nanoparticles interfere with most endotoxin quantification assays. This fact was denoted by the authors of these reports, who overcame the interference, by centrifuging the formulations and measuring the contamination in the supernatant (Grabowski et al., 2016). Unfortunately, due to what was discussed previously, the polymers, and particularly the positively charged, might adsorb the LPS through electrostatic interactions, which means the quantification on the supernatant can be underestimated. Overall, in this example, the conclusions about the mild inflammatory ability of PLGA and PLGA stabilized NPs should be extrapolated with caution, since the use of endotoxin free materials, or the presence of endotoxin inhibitor (i.e., polymycin B) might generate different results.

Genotoxicity

Genotoxicity describes the capacity of the compounds to affect the DNA structure or the cellular apparatus and topoisomerases, modifying the genome fidelity (Słoczynska et al., 2014). Genotoxic effects are not always related with mutations but they can have serious implications for risks of cancer or chronic/heritable diseases (Słoczynska et al., 2014; Lorscheidt and Lamprecht, 2016; Dusinska et al., 2017).

NMs can cause damage to cell's DNA through direct and indirect interactions (Magdolenova et al., 2013; Lorscheidt and Lamprecht, 2016; Dusinska et al., 2017). In fact, upon cellular uptake, NMs might reach the nucleus and contact with cell genetic material, leading to physical or chemical alterations (Magdolenova et al., 2013; Lorscheidt and Lamprecht, 2016; Dusinska et al., 2017). Importantly, this direct interaction is limited by the particle size. Particles ranging between 8 and 10 nm of diameter may reach the nuclear compartment through nuclear pores, whether 15–60 nm particles will only access the nucleus during cellular division when the nuclear wall is disrupted (Barillet et al., 2010). However, indirect interactions have a greater significance for genotoxicity, since several biomolecules involved in normal gene function (i.e., DNA repair) and cell division (i.e., DNA transcription and replication) can interact with even larger NMs, altering its function and consequently leading to DNA injury or chromosome malformation (Lorscheidt and Lamprecht, 2016; Dusinska et al., 2017). For instance, oxidative stress is a key mechanism by which NMs can cause DNA injury (Dusinska et al., 2017). Therefore, data showing non-cytotoxic increase of ROS should imply genotoxicity studies to assess the degree of damage caused by the oxidative stress (Lorscheidt and Lamprecht, 2016).

Several assays are described in the literature for genotoxicity assessment and include in vitro and in vivo approaches. In vitro assays are commonly performed in cell lines, such as the mouse lymphoma L5178Y TK+/− 3.7.2C cells, the TK6 human lymphoblastoid cells and rodent fibroblastic cell lines (CHL-IU, CHO and V79 cells) (Lorge et al., 2016). Regarding in vivo studies, the bacterial reverse mutation test (AMES test) is the most commonly used initial screening performed. Also, the Allium cepa model, allows for a simple and cost-effective assay where DNA damage is assessed after the roots of the plant grow in direct contact with the substance of interest (Bosio and Laughinghouse IV, 2012). Alternatively, other in vivo studies comprise the use of Zebrafish (Danio rerio) due to their molecular and physiological similarities with humans, therefore giving a high-throughput for genotoxicity (Chakravarthy et al., 2014). Rodents and other mammals are also widely used for genotoxicity assessment. In all these models, the comet assay, the micronucleus assay and the chromosome aberrations test are the most common used tests to evaluate nanoparticles toxicity (Magdolenova et al., 2013).

Importantly, some considerations have been published by OECD regarding the protocols to assess genotoxicity of NMs, namely the “2018 Report No. 85—Evaluation of in vitro methods for human hazard assessment applied in the OECD Testing Programme for the Safety of Manufactured Nanomaterials” and “2014 Report No. 43—Genotoxicity of Manufactured Nanomaterials: Report of the OECD expert meeting” (OECD, 2014, 2018a).

Data collected from the literature assessing genotoxicity of polymeric NMs is summarized in Table 6. Again, most of the data collected refers to chitosan and PLGA based NPs and should be carefully analyzed. First, we must recognize we are comparing NPs comprising a particular polymer (chitosan or PLGA) but whose chemical specifications can differ and whose composition and characteristics are very diverse. Also, comparisons should ideally be performed only when the same test is applied. In detail, chitosan/poly(methacrylic acid) NPs induced a concentration dependent genotoxic effect according to the cytogenetic test using human lymphocyte culture (De Lima et al., 2010). However, the same report reported no evidence for DNA alterations using the Allium Cepa assay (De Lima et al., 2010). In another study, Eudragit® S100/alginate enclosed chitosan calcium phosphate-loaded lactoferrin nanocapsules, was considered non-genotoxic based on the Allium Cepa and the comet assay in Vero cells (Leng et al., 2018). Overall, these two studies comprising nanoparticles with chitosan in their composition, presented a different conclusion for the NM genotoxicity, but if we compare only the same assay (Allium Cepa assay), the results were similar. Another interesting fact, is the heterogeneity of results that may be achieved with different cell lines. For instance, Platel et al. used three different cell lines, and three different PLGA NPs and evaluated genotoxicity using the comet assay and the micronucleus test (Platel et al., 2016). For bare PLGA NPs, no genotoxicity effects were verified in none of the 3 cell lines with both tests (Platel et al., 2016). On the other hand, CTAB stabilized PLGA NPs induced an increase in the number of micronuclei only in one of the cell lines (micronucleous test in HBE14o- cells) (Platel et al., 2016). These examples illustrate how an extrapolation based on one single genotoxicity assay (or cellular/animal model) can be misleading.

TABLE 6
www.frontiersin.org

Table 6. Review of original articles assessing the genotoxicity of polymeric nanoparticles according to different testing methodologies.

Toxicity on Reproduction

The extrapolation to human health of toxic effects on reproduction using in vitro and animal models presents several specific limitations, such as the differences in reproductive structures and endocrine functions or the duration of gestation or spermatogenesis period (Das et al., 2016). Also, alike other studies, the tested concentrations and doses are much higher than the clinically relevant doses in humans (Das et al., 2016). Nevertheless, the toxicity on reproduction is a valuable endpoint since it allows the prediction of health effects not only of individuals but also of the next generation (Dusinska et al., 2017).

As mentioned before, toxicity on reproduction might be evaluated using in vitro and in vivo studies. For instance, in vitro assays test the toxicity of nanoparticles in cells from reproductive organs (such as blastocysts and granulosa cells) or use ex vivo placentae or sperm from healthy donors (Ema et al., 2010; Sun et al., 2013; Brohi et al., 2017). In these examples, the authors expect to see direct toxicity of the NPs in reproductive system cells, or to evaluate the ability of the NPs to cross for instance the placental barrier (Ema et al., 2010; Brohi et al., 2017).

Regarding in vivo testing, the use of mice as a mammalian model provides analogous experimental conditions to humans. However, the investigation of early embryonic developmental effects occurring in utero are not easily detectable (Sun et al., 2013). Interestingly, the zebrafish model has been widely applied as a rapid and cost-effective whole animal model to assess reproductive toxicity (Hu et al., 2011). Characteristics like the small size, rapidity to reach sexual maturity, great number of eggs (200–300) and the possibility to examine every stage of embryonic development through its transparency, make zebrafish one of the most used animal models (Wang et al., 2016).

Results from toxicity on reproduction assays with polymeric NMs are summarized in Table 7. The results for chitosan NPs (blend and bare) are consistent between reports. In fact, it appears that chitosan based NPs induce embryonic malformations when directly in contact with embryos, or intravenously administered to animal models (Hu et al., 2011; Park et al., 2013; Choi et al., 2016; Wang et al., 2016; Yostawonkul et al., 2017). However, this effect is not verified in when PLGA NPs coated with chitosan are administered through the oral route in Sprague Dawley rats (Sharma et al., 2017). Though, this conclusion is only speculative. In order to have a proven conclusion, the oral route should be tested for toxicity on reproduction using the same NPs as were used for the intravenous administration and embryonic incubation experiments. Otherwise, we cannot be sure if the result is due to the administration route, or the NPs composition and characteristics. Nevertheless, other study using PLGA based NPs also tested toxicity on reproduction through the in vitro zebrafish embryonic model, and found no toxicity for those nanoparticles (Chen et al., 2017).

TABLE 7
www.frontiersin.org

Table 7. Review of original articles assessing toxicity on reproduction induced by polymeric nanoparticles.

Hemocompatibility

Hemocompatibility is frequently assessed as an endpoint of biocompatibility for chemicals and particularly NMs. In fact, blood is the first target when considering intravenous injections of NMs, but it is also a surrogate target model for other routes of exposure, since its high complexity allows for an approximation the overall body response (Tulinska et al., 2015).

In particular, hemolysis which is associated to red blood cells damage is believed to have a good correlation with toxicity, since the in vitro hemolytic assays show results that greatly relate with in vivo toxicity studies (Dobrovolskaia and McNeil, 2013).

In 2008, Dobrovolskaia et al. published a report describing the validation of an in vitro assay for the analysis of nanoparticle hemolytic properties and main interferences (Dobrovolskaia et al., 2008). In 2013, ASTM International standards organization published the Standard Test Method for Analysis of Hemolytic Properties of Nanoparticles and defined a material as hemolytic if the hemolysis values are above 5% and as moderately hemolytic if they are between 2 and 5% (ASTM International, 2013; Dobrovolskaia and McNeil, 2013). Therefore, the existence of this protocol contributes to the use of standardized procedures among research groups, allowing comparisons and extrapolations of results.

From Table 8 we can acknowledge several authors reporting the hemolytic activity of diverse polymeric NMs. An important remark is the fact that a number of papers describe the hemolytic activity of drug loaded formulations and compare it to the free drug, but not with the unloaded nanocarrier (Essa et al., 2012; Gupta et al., 2012; Altmeyer et al., 2016; Radwan et al., 2017a). These results generally demonstrate a lower hemolysis rate of the drug loaded polymeric NM in comparison to the free drug, but still a significant hemolysis (>5%) (Essa et al., 2012; Gupta et al., 2012; Radwan et al., 2017a). In these situations, no conclusion regarding the hemolytic activity of the polymeric NM itself can be drawn. On the other hand, some other authors, test the unloaded nanoparticles but make no disclosure of their concentration (Altmeyer et al., 2016; Moraes Moreira Carraro et al., 2017).

TABLE 8
www.frontiersin.org

Table 8. Review of original articles assessing hemolysis induced by polymeric nanoparticles.

Nevertheless, polymeric NMs appear to present good hemocompability profile, as in most tested cases, hemolysis is a concentration dependent phenomenon, reaching significant values only for high NM concentrations. Also, the encapsulation of hemolytic drugs in polymeric NMs decreases their hemolytic activity.

Discussion

Most information available on nanotoxicity is related to inorganic NMs, such as zinc oxide NPs, nanoscale silver clusters, and titanium dioxide NPs or carbon nanotubes (Yuan et al., 2015). Information related to polymeric NMs toxicity that could be correlated with their effects on human health is still scarce and poorly harmonized.

The majority of reports on polymeric NMs are focused in optimizing the nanocarrier features, such as size, physical stability and drug loading efficacy, and in performing preliminary cytocompatibility testing (mainly through MTT and LDH assays) and proving effectiveness of the drug loaded formulation, using the most diverse cell lines (Lorscheidt and Lamprecht, 2016). Toxicological studies exploring the biological effects of the polymeric NMs, particularly regarding immune system interaction are often disregarded. Though, as suggested by the safe-by-design concept, the toxicity study of NMs should be the starting point for the formulation development.

After our research on original peer reviewed articles, we selected the following endpoints to analyze that are crucial to understand the toxicity of nanobiomaterials for drug delivery: acute toxicity, repeated-dose toxicity, inflammation, oxidative stress, genotoxicity (including carcinogenicity and mutagenicity) toxicity on reproduction, and hemolysis. Importantly, one of the first conclusions to retain is that among different research groups, the methodologies, the animal or cellular model, the dose or concentration, the assay duration and notably, the polymeric NM properties, are not the same, making it difficult to compare and establish trends. This issue derives in part from the absence of regulatory binding and standardized methodologies and guidelines which hardens the comparison of safety/toxicity assessments in different reports (Dhawan and Sharma, 2010), and ultimately, makes it difficult to extrapolate safety profiles for human health. A similar conclusion was achieved by Park and coworkers, who discussed the status of in vitro toxicity studies for wide-ranging NMs, particularly cytotoxicity, oxidative stress, inflammation and genotoxicity and established that important limitations were preventing their use for human health risk assessment (Park et al., 2009).

Among the different polymeric NMs available, the most studied and reported are chitosan and PLGA nanoparticles. “Chitosan nanoparticles” and “PLGA nanoparticles” are general terms used for an endless number of different nanoparticles comprising multiple polymeric combinations, cross-links and surfactants, and therefore, displaying diverse physical and chemical properties as illustrated by the first 3 columns of Tables 38. As expected, these variables, together with the great diversity of protocols employed by different authors for the same assays, generates ambiguous results that prevent the establishment of trends between the nanocarriers characteristics and the expected toxicological endpoints.

An adequate characterization of the polymeric NMs is crucial for a comprehensive interpretation of the results but also to allow a comparison between different NMs. In 2018, in the context of EU FP-7 GUIDEnano project, it was published the development of a systematic method to assess similarity between NMs that would allow the extrapolation of results for human hazard evaluation purpose (Park et al., 2018). In that methodology they defined the following parameters for assessing similarities between NMs: chemical composition, crystalline form, impurities, primary size distribution, aggregate/agglomerate size distribution, density, and shape. Importantly, those parameters should be tested and compared in relevant media accordingly to the exposure route or toxicity test. However, in the process of developing such methodology, the authors identified several challenges that prevented the establishment of thresholds for establishing similarity. They suggest that the awareness of researchers for the relevance of characterizing NMs when performing hazard assessments is increasing which can lead to the establishment of the thresholds in the future, facilitating the extrapolation of hazard endpoints between similar NMs. Indeed, among the different research articles analyzed, the lack of broad characterization is frequent, sometimes even ignoring important parameters, such as the polymer molecular weight or the nanoparticle size.

Another aspect that should be taken into consideration when characterizing the polymeric NMs to study their biological effects is the endotoxin contamination. In fact, when discussing for instance cytokine stimulation or oxidative stress, endotoxin contamination should not be neglected. Nevertheless, endotoxin quantification (or its acknowledgment) on chitosan and other polymeric NMs is still scarce, which compromises some of the results found in the literature regarding their bioactivity and toxicity. In addition, despite testing the presence of endotoxins is a common procedure in laboratory and several commercial tests are available, they need to be validated for use with NMs, since most are based on optical assays and may be affected by the optical density of NPs (Dobrovolskaia et al., 2010).

Not only endotoxin detection assays are susceptible of interference from NMs and consequently misinterpretation of the results. Therefore, one way of trying to overcome this problem is to use different assays to evaluate the same endpoint. Additionally, experiment controls, such as the incubation of probes (without biological matrixes) and positive controls with NMs, can reveal whether these NMs might be generating false positive or negative results.

The obstacles identified in this review prevent the identification of toxicity trends and the generation of a useful database where we can rely for the Safe-by-Design. Only by performing in vitro and in vivo harmonized toxicity studies using unloaded polymeric NMs, extensively characterized regarding their intrinsic and extrinsic properties and by performing all necessary controls it is possible to generate such database. At the present time, taking everything into account, the human health risk assessment of polymeric NMs is still dependent on a case-by-case evaluation, and it should comprise the evaluation of parameters, such as the route of administration and dose, among others, to define the required tests for the hazard assessment (i.e., type of in vitro and in vivo studies).

Author Contributions

SJ gathered the information, analyzed, and wrote the first draft of the manuscript. SJ, MS, CS, GB, PW, and OB defined the subjects for discussion. All authors contributed to manuscript revision, read, and approved the submitted version.

Funding

This work was financed by the European Regional Development Fund (ERDF), through the Centro 2020 Regional Operational Programme under project CENTRO-01-0145-FEDER-00008:BrainHealth 2020, and through the COMPETE 2020—Operational Programme for Competitiveness and Internationalization and Portuguese national funds via FCT—Fundação para a Ciência e a Tecnologia, I.P., under project PROSAFE/0001/2016, and the strategic projects POCI-01-0145-FEDER-030331 and POCI-01-0145-FEDER-007440 (UID/NEU/04539/2019). This work is part of the GoNanoBioMat project and has received funding from the Horizon 2020 framework program of the European Union, ProSafe Joint Transnational Call 2016 and from the CTI (1.1.2018 Innosuisse), under grant agreement Number 19267.1 PFNM-NM.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Footnotes

References

Agarwal, S., Piesco, N. P., Johns, L. P., and Riccelli, A. E. (1995). Differential expression of IL-1 beta, TNF-alpha, IL-6, and IL-8 in human monocytes in response to lipopolysaccharides from different microbes. J. Dent. Res. 74, 1057–1065. doi: 10.1177/00220345950740040501

PubMed Abstract | CrossRef Full Text | Google Scholar

Agrawal, U., Sharma, R., Gupta, M., and Vyas, S. P. (2014). Is nanotechnology a boon for oral drug delivery? Drug Discov. Today 19, 1530–1546. doi: 10.1016/j.drudis.2014.04.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Ai, J., Biazar, E., Jafarpour, M., Montazeri, M., Majdi, A., Aminifard, S., et al. (2011). Nanotoxicology and nanoparticle safety in biomedical designs. Int. J. Nanomedicine 1117–1127. doi: 10.2147/IJN.S16603

PubMed Abstract | CrossRef Full Text | Google Scholar

Altmeyer, C., Karam, T. K., Khalil, N. M., and Mainardes, R. M. (2016). Tamoxifen-loaded poly(L-lactide) nanoparticles: development, characterization and in vitro evaluation of cytotoxicity. Mater. Sci. Eng. C 60, 135–142. doi: 10.1016/j.msec.2015.11.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Aluani, D., Tzankova, V., Kondeva-Burdina, M., Yordanov, Y., Nikolova, E., Odzhakov, F., et al. (2017). Evaluation of biocompatibility and antioxidant efficiency of chitosan-alginate nanoparticles loaded with quercetin. Int. J. Biol. Macromol. 103, 771–782. doi: 10.1016/j.ijbiomac.2017.05.062

CrossRef Full Text | Google Scholar

Aragao-Santiago, L., Hillaireau, H., Grabowski, N., Mura, S., Nascimento, T. L., Dufort, S., et al. (2015). Compared in vivo toxicity in mice of lung delivered biodegradable and non-biodegradable nanoparticles. Nanotoxicology 10, 292–302. doi: 10.3109/17435390.2015.1054908

PubMed Abstract | CrossRef Full Text | Google Scholar

Aranda, A., Sequedo, L., Tolosa, L., Quintas, G., Burello, E., Castell, J. V., et al. (2013). Dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay: a quantitative method for oxidative stress assessment of nanoparticle-treated cells. Toxicol. in vitro 27, 954–963. doi: 10.1016/j.tiv.2013.01.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Arora, D., Dhanwal, V., Nayak, D., Saneja, A., Amin, H., ur Rasool, R., et al. (2016). Preparation, characterization and toxicological investigation of copper loaded chitosan nanoparticles in human embryonic kidney HEK-293 cells. Mater. Sci. Eng. C 61, 227–234. doi: 10.1016/j.msec.2015.12.035

PubMed Abstract | CrossRef Full Text | Google Scholar

ASTM International (2013). ASTM E2524-08(2013)–Standard Test Method for Analysis of Hemolytic Properties of Nanoparticles.

Banik, B. L., Fattahi, P., and Brown, J. L. (2016). Polymeric nanoparticles: the future of nanomedicine. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 8, 271–299. doi: 10.1002/wnan.1364

PubMed Abstract | CrossRef Full Text | Google Scholar

Barillet, S., Jugan, M. L., Laye, M., Leconte, Y., Herlin-Boime, N., Reynaud, C., et al. (2010). In vitro evaluation of SiC nanoparticles impact on A549 pulmonary cells: Cyto-, genotoxicity and oxidative stress. Toxicol. Lett. 198, 324–330. doi: 10.1016/j.toxlet.2010.07.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Barratt, G. M. (2000). Therapeutic applications of colloidal drug carriers. Pharm. Sci. Technol. Today 3, 163–171. doi: 10.1016/S1461-5347(00)00255-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Bazile, D., Prud'homme, C., Bassoullet, M. T., Marlard, M., Spenlehauer, G., and Veillard, M. (1995). Stealth Me.PEG-PLA nanoparticles avoid uptake by the mononuclear phagocytes system. J. Pharm. Sci. 84, 493–498. doi: 10.1002/jps.2600840420

PubMed Abstract | CrossRef Full Text | Google Scholar

Behzadi, S., Serpooshan, V., Sakhtianchi, R., Muller, B., Landfester, K., Crespy, D., et al. (2014). Protein corona change the drug release profile of nanocarriers: the “overlooked” factor at the nanobio interface. Colloids Surf. B Biointerfaces 123, 143–149. doi: 10.1016/j.colsurfb.2014.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Behzadi, S., Serpooshan, V., Tao, W., Hamaly, M. A., Alkawareek, M. Y., Dreaden, E. C., et al. (2017). Cellular uptake of nanoparticles: journey inside the cell. Chem. Soc. Rev. 46, 4218–4244. doi: 10.1039/C6CS00636A

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhatia, S. (2016). “Nanoparticles types, classification, characterization, fabrication methods and drug delivery applications,” in Natural Polymer Drug Delivery Systems (Cham: Springer), 33–93.

Google Scholar

Blanco, E., Shen, H., and Ferrari, M. (2015). Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat. Biotechnol. 33, 941–951. doi: 10.1038/nbt.3330

PubMed Abstract | CrossRef Full Text | Google Scholar

Bor, G., Mytych, J., Zebrowski, J., Wnuk, M., and Sanli-Mohamed, G. (2016). Cytotoxic and cytostatic side effects of chitosan nanoparticles as a non-viral gene carrier. Int. J. Pharm. 513, 431–437. doi: 10.1016/j.ijpharm.2016.09.058

PubMed Abstract | CrossRef Full Text | Google Scholar

Bosio, S., and Laughinghouse IV, H. D. (2012). “Bioindicator of genotoxicity: the Allium cepa test,” in Environmental Contamination, ed J. Srivastava (IntechOpen).

Google Scholar

Boyes, W. K., Thornton, B. L. M., Al-Abed, S. R., Andersen, C. P., Bouchard, D. C., Burgess, R. M., et al. (2017). A comprehensive framework for evaluating the environmental health and safety implications of engineered nanomaterials. Crit. Rev. Toxicol. 47, 767–810. doi: 10.1080/10408444.2017.1328400

PubMed Abstract | CrossRef Full Text | Google Scholar

Brohi, R. D., Wang, L., Talpur, H. S., Wu, D., Khan, F. A., Bhattarai, D., et al. (2017). Toxicity of nanoparticles on the reproductive system in animal models: a review. Front. Pharmacol. 8:606. doi: 10.3389/fphar.2017.00606

PubMed Abstract | CrossRef Full Text | Google Scholar

Buzea, C., Pacheco, I. I., and Robbie, K. (2007). Nanomaterials and nanoparticles: sources and toxicity. Biointerphases 2, MR17–MR71. doi: 10.1116/1.2815690

PubMed Abstract | CrossRef Full Text | Google Scholar

Chakravarthy, S., Sadagopan, S., Nair, A., and Sukumaran, S. K. (2014). Zebrafish as an in vivo high-throughput model for genotoxicity. Zebrafish 11, 154–166. doi: 10.1089/zeb.2013.0924

PubMed Abstract | CrossRef Full Text | Google Scholar

Chang, Y.-M., Lee, Y.-J., Liao, J.-W., Jhan, J.-K., Chang, C.-T., and Chung, Y.-C. (2014). In vitro and in vivo safety evaluation of low molecular weight chitosans prepared by hydrolyzing crab shell chitosans with bamboo shoots chitosanase. Food Chem. Toxicol. 71, 10–16. doi: 10.1016/j.fct.2014.05.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J., Wu, Q., Luo, L., Wang, Y., Zhong, Y., Dai, H., et al. (2017). Dual tumor-targeted poly(lactic-co-glycolic acid)–polyethylene glycol–folic acid nanoparticles: a novel biodegradable nanocarrier for secure and efficient antitumor drug delivery. Int. J. Nanomed. 12, 5745–5760. doi: 10.2147/IJN.S136488

PubMed Abstract | CrossRef Full Text | Google Scholar

Choi, Y.-J., Gurunathan, S., Kim, D., Jang, H. S., Park, W.-J., Cho, S.-G., et al. (2016). Rapamycin ameliorates chitosan nanoparticle-induced developmental defects of preimplantation embryos in mice. Oncotarget 7, 74658–74677. doi: 10.18632/oncotarget.10813

PubMed Abstract | CrossRef Full Text | Google Scholar

Da Silva, J., Jesus, S., Bernardi, N., Colaço, M., and Borges, O. (2019). Poly(D,L-lactic acid) nanoparticle size reduction increases its immunotoxicity. Front. Bioeng. Biotechnol. 7:137. doi: 10.3389/fbioe.2019.00137

PubMed Abstract | CrossRef Full Text | Google Scholar

Das, J., Choi, Y.-J., Song, H., and Kim, J.-H. (2016). Potential toxicity of engineered nanoparticles in mammalian germ cells and developing embryos: treatment strategies and anticipated applications of nanoparticles in gene delivery. Hum. Reprod. Update 22, 588–619. doi: 10.1093/humupd/dmw020

PubMed Abstract | CrossRef Full Text | Google Scholar

Date, A. A., Hanes, J., and Ensign, L. M. (2016). Nanoparticles for oral delivery: Design, evaluation and state-of-the-art. J. Control. Release 240, 504–526. doi: 10.1016/j.jconrel.2016.06.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Davydova, V. N., Yermak, I. M., Gorbach, V. I., Krasikova, I. N., and Solov'eva, T. F. (2000). Interaction of bacterial endotoxins with chitosan. Effect of endotoxin structure, chitosan molecular mass, and ionic strength of the solution on the formation of the complex. Biochemistry 65, 1082–1090.

PubMed Abstract | Google Scholar

De Lima, R., Feitosa, L., Pereira A, D. E. S., De Moura, M. R., Aouada, F. A., Mattoso, L. H. C., et al. (2010). Evaluation of the genotoxicity of chitosan nanoparticles for use in food packaging films. J. Food Sci. 75, N89–N96. doi: 10.1111/j.1750-3841.2010.01682.x

PubMed Abstract | CrossRef Full Text | Google Scholar

de Salamanca, A. E. D, Diebold, Y., Calonge, M., Garci'a-Vazquez, C., Callejo, S., Vila, A., et al. (2006). Chitosan nanoparticles as a potential drug delivery system for the ocular surface: toxicity, uptake mechanism and in vivo tolerance. Investig. Opthalmol. Vis. Sci. 47, 1416–1425. doi: 10.1167/iovs.05-0495

CrossRef Full Text | Google Scholar

DeLoid, G., Cohen, J. M., Darrah, T., Derk, R., Rojanasakul, L., Pyrgiotakis, G., et al. (2014). Estimating the effective density of engineered nanomaterials for in vitro dosimetry. Nat. Commun. 5:3514. doi: 10.1038/ncomms4514

PubMed Abstract | CrossRef Full Text | Google Scholar

Dhawan, A., and Sharma, V. (2010). Toxicity assessment of nanomaterials: methods and challenges. Anal. Bioanal. Chem. 398, 589–605. doi: 10.1007/s00216-010-3996-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Dobrovolskaia, M. A., Clogston, J. D., Neun, B. W., Hall, J. B., Patri, A. K., and McNeil, S. E. (2008). Method for analysis of nanoparticle hemolytic properties in vitro. Nano Lett. 8, 2180–2187. doi: 10.1021/nl0805615

PubMed Abstract | CrossRef Full Text | Google Scholar

Dobrovolskaia, M. A., Germolec, D. R., and Weaver, J. L. (2009). Evaluation of nanoparticle immunotoxicity. Nat. Nanotechnol. 4, 411–414. doi: 10.1038/nnano.2009.175

PubMed Abstract | CrossRef Full Text | Google Scholar

Dobrovolskaia, M. A., and McNeil, S. E. (2013). Understanding the correlation between in vitro and in vivo immunotoxicity tests for nanomedicines. J. Control. Release 172, 456–466. doi: 10.1016/j.jconrel.2013.05.025

PubMed Abstract | CrossRef Full Text | Google Scholar

Dobrovolskaia, M. A., Neun, B. W., Clogston, J. D., Ding, H., Ljubimova, J., and McNeil, S. E. (2010). Ambiguities in applying traditional Limulus amebocyte lysate tests to quantify endotoxin in nanoparticle formulations. Nanomedicine 5, 555–562. doi: 10.2217/nnm.10.29

PubMed Abstract | CrossRef Full Text | Google Scholar

Dusinska, M., Tulinska, J., El Yamani, N., Kuricova, M., Liskova, A., Rollerova, E., et al. (2017). Immunotoxicity, genotoxicity and epigenetic toxicity of nanomaterials: new strategies for toxicity testing? Food Chem. Toxicol. 109(Pt 1), 797–811. doi: 10.1016/j.fct.2017.08.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Egusquiaguirre, S. P., Pedraz, J. L., Hernández, R. M., and Igartua, M. (2016). “Nanotherapeutic platforms for cancer treatment: from preclinical development to clinical application,” in Nanoarchitectonics for Smart Delivery and Drug Targeting, eds A.-M. Holban, A. Mihai (Oxford: William Andrew) 813–869. doi: 10.1016/B978-0-323-47347-7.00029-X

CrossRef Full Text | Google Scholar

Ema, M., Kobayashi, N., Naya, M., Hanai, S., and Nakanishi, J. (2010). Reproductive and developmental toxicity studies of manufactured nanomaterials. Reproduct. Toxicol. 30, 343–352. doi: 10.1016/j.reprotox.2010.06.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Essa, S., Louhichi, F., Raymond, M., and Hildgen, P. (2012). Improved antifungal activity of itraconazole-loaded PEG/PLA nanoparticles. J. Microencapsul. 30, 205–217. doi: 10.3109/02652048.2012.714410

PubMed Abstract | CrossRef Full Text | Google Scholar

European Medicines Agency (2006). EMEA/CHMP/79769/2006–Reflection Paper on Nanotechnology-Based Medicinal Products for Human Use.

Fasehee, H., Dinarvand, R., Ghavamzadeh, A., Esfandyari-Manesh, M., Moradian, H., Faghihi, S., et al. (2016). Delivery of disulfiram into breast cancer cells using folate-receptor-targeted PLGA-PEG nanoparticles: in vitro and in vivo investigations. J. Nanobiotechnol. 14:32. doi: 10.1186/s12951-016-0183-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Fernandes, J. C., Eaton, P., Nascimento, H., Gião, M. S., Ramos, Ó. S., Belo, L., et al. (2010). Antioxidant activity of chitooligosaccharides upon two biological systems: Erythrocytes and bacteriophages. Carbohydr. Polym. 79, 1101–1106. doi: 10.1016/j.carbpol.2009.10.050

CrossRef Full Text | Google Scholar

Food and Drug Aministration (2014). Considering Whether an FDA-Regulated Product Involves the Application of Nanotechnology: Guidance for Industry. Food and Drug Aministration (FDA).

Fröhlich, E. (2012). The role of surface charge in cellular uptake and cytotoxicity of medical nanoparticles. Int. J. Nanomedicine 7, 5577–5591. doi: 10.2147/IJN.S36111

PubMed Abstract | CrossRef Full Text | Google Scholar

Gatoo, M. A., Naseem, S., Arfat, M. Y., Mahmood Dar, A., Qasim, K., and Zubair, S. (2014). Physicochemical properties of nanomaterials: implication in associated toxic manifestations. Biomed Res. Int. 2014:498420. doi: 10.1155/2014/498420

PubMed Abstract | CrossRef Full Text | Google Scholar

Gatto, F., and Bardi, G. (2018). Metallic nanoparticles: general research approaches to immunological characterization. Nanomaterials 8:E753. doi: 10.3390/nano8100753

PubMed Abstract | CrossRef Full Text | Google Scholar

Ge, C. C., Du, J. F., Zhao, L. N., Wang, L. M., Liu, Y., Li, D. H., et al. (2011). Binding of blood proteins to carbon nanotubes reduces cytotoxicity. Proc. Natl. Acad. Sci. U.S.A. 108, 16968–16973. doi: 10.1073/pnas.1105270108

PubMed Abstract | CrossRef Full Text | Google Scholar

Grabowski, N., Hillaireau, H., Vergnaud, J., Tsapis, N., Pallardy, M., Kerdine-Romer, S., et al. (2015). Surface coating mediates the toxicity of polymeric nanoparticles towards human-like macrophages. Int. J. Pharm. 482, 75–83. doi: 10.1016/j.ijpharm.2014.11.042

PubMed Abstract | CrossRef Full Text | Google Scholar

Grabowski, N., Hillaireau, H., Vergnaud-Gauduchon, J., Nicolas, V., Tsapis, N., Kerdine-Römer, S., et al. (2016). Surface-modified biodegradable nanoparticles' impact on cytotoxicity and inflammation response on a co-culture of lung epithelial cells and human-like macrophages. J. Biomed. Nanotechnol. 12, 135–146. doi: 10.1166/jbn.2016.2126

PubMed Abstract | CrossRef Full Text | Google Scholar

Guadagnini, R., Halamoda Kenzaoui, B., Walker, L., Pojana, G., Magdolenova, Z., Bilanicova, D., et al. (2013a). Toxicity screenings of nanomaterials: challenges due to interference with assay processes and components of classic in vitro tests. Nanotoxicology 9, 13–24. doi: 10.3109/17435390.2013.829590

PubMed Abstract | CrossRef Full Text | Google Scholar

Guadagnini, R., Moreau, K., Hussain, S., Marano, F., and Boland, S. (2013b). Toxicity evaluation of engineered nanoparticles for medical applications using pulmonary epithelial cells. Nanotoxicology 9, 25–32. doi: 10.3109/17435390.2013.855830

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, L., Chen, B., Liu, R., Liu, P., Xia, G., Wang, Y., et al. (2015). Biocompatibility assessment of polyethylene glycol-poly L-lysine-poly lactic-co-glycolic acid nanoparticles in vitro and in vivo. J. Nanosci. Nanotechnol. 15, 3710–3719. doi: 10.1166/jnn.2015.9509

PubMed Abstract | CrossRef Full Text | Google Scholar

Gupta, S., Agarwal, A., Gupta, N. K., Saraogi, G., Agrawal, H., and Agrawal, G. P. (2012). Galactose decorated PLGA nanoparticles for hepatic delivery of acyclovir. Drug Develop. Industr. Pharm. 39, 1866–1873. doi: 10.3109/03639045.2012.662510

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, H. D., Byeon, Y., Jang, J.-H., Jeon, H. N., Kim, G. H., Kim, M. G., et al. (2016). In vivo stepwise immunomodulation using chitosan nanoparticles as a platform nanotechnology for cancer immunotherapy. Sci. Rep. 6:38348. doi: 10.1038/srep38348

PubMed Abstract | CrossRef Full Text | Google Scholar

Hinderliter, P. M., Minard, K. R., Orr, G., Chrisler, W. B., Thrall, B. D., Pounds, J. G., et al. (2010). ISDD: a computational model of particle sedimentation, diffusion and target cell dosimetry for in vitro toxicity studies. Part. Fibre Toxicol. 7:36. doi: 10.1186/1743-8977-7-36

PubMed Abstract | CrossRef Full Text | Google Scholar

Hoshyar, N., Gray, S., Han, H., and Bao, G. (2016). The effect of nanoparticle size on in vivo pharmacokinetics and cellular interaction. Nanomedicine 11, 673–692. doi: 10.2217/nnm.16.5

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, Y., Wang, H., and Gao, J. (2011). Toxicity evaluation of biodegradable chitosan nanoparticles using a zebrafish embryo model. Int. J. Nanomedicine 6, 3351–339. doi: 10.2147/IJN.S25853

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, Y., Gao, H., Gou, M., Ye, H., Liu, Y., Gao, Y., et al. (2010). Acute toxicity and genotoxicity studies on poly(ε-caprolactone)-poly(ethylene glycol)-poly(ε-caprolactone) nanomaterials. Mutat. Res. Genet. Toxicol. Environ. Mutage. 696, 101–106. doi: 10.1016/j.mrgentox.2009.12.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Je, J.-Y., Park, P.-J., and Kim, S.-K. (2004). Free radical scavenging properties of hetero-chitooligosaccharides using an ESR spectroscopy. Food Chem. Toxicol. 42, 381–387. doi: 10.1016/j.fct.2003.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Jena, S. K., and Sangamwar, A. T. (2016). Polymeric micelles of amphiphilic graft copolymer of α -tocopherol succinate- g -carboxymethyl chitosan for tamoxifen delivery: synthesis, characterization and in vivo pharmacokinetic study. Carbohydr. Polym. 151, 1162–1174. doi: 10.1016/j.carbpol.2016.06.078

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiménez, A. S., MacCalman, L., Belut, E., Goekce, S., Sirokova, L., Cowie, H., et al. (2016). NANoREG Deliverable 3.08–Improved and Validated Occupational Exposure Models of Release, Exposure, Dispersion and Transfer. National Institute for Public Health and the Environment Ministry of Health, Welfare and Sport

Jindal, A. B. (2017). The effect of particle shape on cellular interaction and drug delivery applications of micro- and nanoparticles. Int. J. Pharm. 532, 450–465. doi: 10.1016/j.ijpharm.2017.09.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalyanaraman, B., Darley-Usmar, V., Davies, K. J. A., Dennery, P. A., Forman, H. J., Grisham, M. B., et al. (2012). Measuring reactive oxygen and nitrogen species with fluorescent probes: challenges and limitations. Free Radical Biol. Med. 52, 1–6. doi: 10.1016/j.freeradbiomed.2011.09.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Kanwal, Z., Raza, M., Manzoor, F., Riaz, S., Jabeen, G., Fatima, S., et al. (2019). A comparative assessment of nanotoxicity induced by metal (silver, nickel) and metal oxide (cobalt, chromium) nanoparticles in Labeo rohita. Nanomaterials 9:309. doi: 10.3390/nano9020309

PubMed Abstract | CrossRef Full Text | Google Scholar

Khan, H. A., and Shanker, R. (2015). Toxicity of nanomaterials. Biomed Res. Int. 2015:521014. doi: 10.1155/2015/521014

PubMed Abstract | CrossRef Full Text | Google Scholar

Khang, D., Lee, Y. K., Choi, E.-J., Webster, T. J., and Kim, S.-H. (2014). Effect of the protein corona on nanoparticles for modulating cytotoxicity and immunotoxicity. Int. J. Nanomedicine 10, 97–113. doi: 10.2147/IJN.S72998

PubMed Abstract | CrossRef Full Text | Google Scholar

Khanna, P., Ong, C., Bay, B., and Baeg, G. (2015). Nanotoxicity: an interplay of oxidative stress, inflammation and cell death. Nanomaterials 5, 1163–1180. doi: 10.3390/nano5031163

PubMed Abstract | CrossRef Full Text | Google Scholar

Kononenko, V., Narat, M., and Drobne, D. (2015). Nanoparticle interaction with the immune system/Interakcije nanodelcev z imunskim sistemom. Arch. Industr. Hyg. Toxicol. 66, 97–108. doi: 10.1515/aiht-2015-66-2582

CrossRef Full Text | Google Scholar

Koppolu, B., and Zaharoff, D. A. (2013). The effect of antigen encapsulation in chitosan particles on uptake, activation and presentation by antigen presenting cells. Biomaterials 34, 2359–2369. doi: 10.1016/j.biomaterials.2012.11.066

PubMed Abstract | CrossRef Full Text | Google Scholar

Kroll, A., Pillukat, M. H., Hahn, D., and Schnekenburger, J. (2012). Interference of engineered nanoparticles with in vitro toxicity assays. Arch. Toxicol. 86, 1123–1136. doi: 10.1007/s00204-012-0837-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, V., Leekha, A., Tyagi, A., Kaul, A., Mishra, A. K., and Verma, A. K. (2017). Preparation and evaluation of biopolymeric nanoparticles as drug delivery system in effective treatment of rheumatoid arthritis. Pharm. Res. 34, 654–667. doi: 10.1007/s11095-016-2094-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Landsiedel, R., Fabian, E., Ma-Hock, L., Wohlleben, W., Wiench, K., Oesch, F., et al. (2012). Toxico-/biokinetics of nanomaterials. Arch. Toxicol. 86, 1021–1060. doi: 10.1007/s00204-012-0858-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Landsiedel, R., Ma-Hock, L., Wiench, K., Wohlleben, W., and Sauer, U. G. (2017). Safety assessment of nanomaterials using an advanced decision-making framework, the DF4nanoGrouping. J. Nanopart. Res. 19:171. doi: 10.1007/s11051-017-3850-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Legaz, S., Exposito, J.-Y., Lethias, C., Viginier, B., Terzian, C., and Verrier, B. (2016). Evaluation of polylactic acid nanoparticles safety using Drosophila model. Nanotoxicology 10, 1136–1143. doi: 10.1080/17435390.2016.1181806

PubMed Abstract | CrossRef Full Text | Google Scholar

Leng, K. M., Vijayarathna, S., Jothy, S. L., Sasidharan, S., and Kanwar, J. R. (2018). In vitro and in vivo toxicity assessment of alginate/eudragit S 100-enclosed chitosan–calcium phosphate-loaded iron saturated bovine lactoferrin nanocapsules (Fe-bLf NCs). Biomed. Pharmacother. 97, 26–37. doi: 10.1016/j.biopha.2017.10.121

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, C., Shen, Y., Sun, C., Nihad, C., and Tu, J. (2014). Immunosafety and chronic toxicity evaluation of monomethoxypoly(ethylene glycol)-b-poly(lactic acid) polymer micelles for paclitaxel delivery. Drug Deliv. 23, 888–895. doi: 10.3109/10717544.2014.971196

PubMed Abstract | CrossRef Full Text | Google Scholar

Lieder, R., Gaware, V. S., Thormodsson, F., Einarsson, J. M., Ng, C.-H., Gislason, J., et al. (2013). Endotoxins affect bioactivity of chitosan derivatives in cultures of bone marrow-derived human mesenchymal stem cells. Acta Biomater. 9, 4771–4778. doi: 10.1016/j.actbio.2012.08.043

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., Kong, M., Feng, C., Yang, K. K., Li, Y., Su, J., et al. (2013). Biocompatibility, cellular uptake and biodistribution of the polymeric amphiphilic nanoparticles as oral drug carriers. Colloids Surfaces B 103, 345–353. doi: 10.1016/j.colsurfb.2012.11.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Lorge, E., Moore, M. M., Clements, J., O'Donovan, M., Fellows, M. D., Honma, M., et al. (2016). Standardized cell sources and recommendations for good cell culture practices in genotoxicity testing. Mutat. Res. Genet. Toxicol. Environ. Mutage. 809, 1–15. doi: 10.1016/j.mrgentox.2016.08.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Lorscheidt, S., and Lamprecht, A. (2016). Safety assessment of nanoparticles for drug delivery by means of classic in vitro assays and beyond. Expert Opin. Drug Deliv. 13, 1545–1558. doi: 10.1080/17425247.2016.1198773

PubMed Abstract | CrossRef Full Text | Google Scholar

Lü, J.-M., Lin, P. H., Yao, Q., and Chen, C. (2010). Chemical and molecular mechanisms of antioxidants: experimental approaches and model systems. J. Cell. Mol. Med. 14, 840–860. doi: 10.1111/j.1582-4934.2009.00897.x,

PubMed Abstract | CrossRef Full Text | Google Scholar

Lü, X., Zheng, B., Tang, X., Zhao, L., Lu, J., Zhang, Z., et al. (2011). In vitro biomechanical and biocompatible evaluation of natural hydroxyapatite/chitosan composite for bone repair. J. Appl. Biomater. Biomech. 9, 11–18. doi: 10.5301/JABB.2011.6474

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, N., Ma, C., Li, C., Wang, T., Tang, Y., Wang, H., et al. (2013). Influence of nanoparticle shape, size, and surface functionalization on cellular uptake. J. Nanosci. Nanotechnol. 13, 6485–6498. doi: 10.1166/jnn.2013.7525

PubMed Abstract | CrossRef Full Text | Google Scholar

Machado, R. L., de Arruda, E. J., Santana, C. C., and Bueno, S. M. A. (2006). Evaluation of a chitosan membrane for removal of endotoxin from human IgG solutions. Process Biochem. 41, 2252–2257. doi: 10.1016/j.procbio.2006.05.015

CrossRef Full Text | Google Scholar

Magalhaes, P. O., Lopes, A. M., Mazzola, P. G., Rangel-Yagui, C., Penna, T. C., Pessoa, A., et al. (2007). Methods of endotoxin removal from biological preparations: a review. J. Pharm. Pharm. Sci. 10, 388–404.

PubMed Abstract | Google Scholar

Magdolenova, Z., Collins, A., Kumar, A., Dhawan, A., Stone, V., and Dusinska, M. (2013). Mechanisms of genotoxicity. A review of in vitro and in vivo studies with engineered nanoparticles. Nanotoxicology 8, 233–278. doi: 10.3109/17435390.2013.773464

PubMed Abstract | CrossRef Full Text | Google Scholar

Maity, S., Mukhopadhyay, P., Kundu, P. P., and Chakraborti, A. S. (2017). Alginate coated chitosan core-shell nanoparticles for efficient oral delivery of naringenin in diabetic animals—an in vitro and in vivo approach. Carbohydr. Polym. 170, 124–132. doi: 10.1016/j.carbpol.2017.04.066

PubMed Abstract | CrossRef Full Text | Google Scholar

Moraes Moreira Carraro, T. C., Altmeyer, C., Maissar Khalil, N., and Mara Mainardes, R. (2017). Assessment of in vitro antifungal efficacy and in vivo toxicity of Amphotericin B-loaded PLGA and PLGA-PEG blend nanoparticles. J. Mycol. Méd. 27, 519–529. doi: 10.1016/j.mycmed.2017.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Moritz, M., and Geszke-Moritz, M. (2015). Recent developments in application of polymeric nanoparticles as drug carriers. Adv. Clin. Exper. Med. 24, 749–758. doi: 10.17219/acem/31802

PubMed Abstract | CrossRef Full Text | Google Scholar

Mukhopadhyay, P., Chakraborty, S., Bhattacharya, S., Mishra, R., and Kundu, P. P. (2015). pH-sensitive chitosan/alginate core-shell nanoparticles for efficient and safe oral insulin delivery. Int. J. Biol. Macromol. 72, 640–648. doi: 10.1016/j.ijbiomac.2014.08.040

PubMed Abstract | CrossRef Full Text | Google Scholar

Nadesh, R., Narayanan, D., PR, S., Vadakumpully, S., Mony, U., Koyakkutty, M., et al. (2013). Hematotoxicological analysis of surface-modified and -unmodified chitosan nanoparticles. J. Biomed. Mater. Res. Part A 101, 2957–2966. doi: 10.1002/jbm.a.34591

PubMed Abstract | CrossRef Full Text | Google Scholar

Ngo, D.-H., and Kim, S.-K. (2014). Antioxidant effects of chitin, chitosan, and their derivatives. Adv. Food Nutr. Res. 73, 15–31. doi: 10.1016/B978-0-12-800268-1.00002-0

PubMed Abstract | CrossRef Full Text | Google Scholar

OECD (2014). “Genotoxicity of manufactured nanomaterials: report of the OECD expert meeting,” in Series on the Safety of Manufactured Nanomaterials No. 43 (Paris: OECD Environment, Health and Safety Publications).

OECD (2018a). “Evaluation of in vitro methods for human hazard assessment applied in the OECD Testing Programme for the Safety of Manufactured Nanomaterials,” in Series on the Safety of Manufactured Nanomaterials No. 85 (Paris: OECD Environment, Health and Safety Publications).

OECD (2018b). “Test No. 412: subacute inhalation toxicity: 28-day study,” in OECD Guidelines for the Testing of Chemicals, Section 4: Health Effects (Paris: OECD Publishing).

OECD (2018c). “Test No. 413: subchronic inhalation toxicity: 90-day study,” in OECD Guidelines for the Testing of Chemicals, Section 4: Health Effects (Paris: OECD Publishing).

Omar Zaki, S. S., Katas, H., and Hamid, Z. A. (2015). Lineage-related and particle size-dependent cytotoxicity of chitosan nanoparticles on mouse bone marrow-derived hematopoietic stem and progenitor cells. Food Chem. Toxicol. 85, 31–44. doi: 10.1016/j.fct.2015.05.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Oostingh, G. J., Casals, E., Italiani, P., Colognato, R., Stritzinger, R., Ponti, J., et al. (2011). Problems and challenges in the development and validation of human cell-based assays to determine nanoparticle-induced immunomodulatory effects. Part. Fibre Toxicol. 8:8. doi: 10.1186/1743-8977-8-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Oparka, M., Walczak, J., Malinska, D., van Oppen, L. M. P. E., Szczepanowska, J., Koopman, W. J. H., et al. (2016). Quantifying ROS levels using CM-H 2 DCFDA and HyPer. Methods 109, 3–11. doi: 10.1016/j.ymeth.2016.06.008

CrossRef Full Text | Google Scholar

Palmer, B., and DeLouise, L. (2016). Nanoparticle-enabled transdermal drug delivery systems for enhanced dose control and tissue targeting. Molecules 21:E1719. doi: 10.3390/molecules21121719

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, M.-R., Gurunathan, S., Choi, Y.-J., Kwon, D.-N., Han, J.-W., Cho, S.-G., et al. (2013). Chitosan nanoparticles cause pre- and post-implantation embryo complications in mice. Biol. Reprod. 88:88. doi: 10.1095/biolreprod.112.107532

CrossRef Full Text | Google Scholar

Park, M. V. D. Z., Catalán, J., Ferraz, N., Cabellos, J., Vanhauten, R., Vázquez-Campos, S., et al. (2018). Development of a systematic method to assess similarity between nanomaterials for human hazard evaluation purposes–lessons learnt. Nanotoxicology 12, 652–676. doi: 10.1080/17435390.2018.1465142

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, M. V. D. Z., Lankveld, D. P. K., van Loveren, H., and de Jong, W. H. (2009). The status of in vitro toxicity studies in the risk assessment of nanomaterials. Nanomedicine 4, 669–685. doi: 10.2217/nnm.09.40

PubMed Abstract | CrossRef Full Text | Google Scholar

Pillai, G. J., Greeshma, M. M., and Menon, D. (2015). Impact of poly(lactic-co-glycolic acid) nanoparticle surface charge on protein, cellular and haematological interactions. Colloids Surf. B Biointerfaces 136, 1058–1066. doi: 10.1016/j.colsurfb.2015.10.047

PubMed Abstract | CrossRef Full Text | Google Scholar

Platel, A., Carpentier, R., Becart, E., Mordacq, G., Betbeder, D., and Nesslany, F. (2016). Influence of the surface charge of PLGA nanoparticles on their in vitro genotoxicity, cytotoxicity, ROS production and endocytosis. J. Appl. Toxicol. 36, 434–444. doi: 10.1002/jat.3247

PubMed Abstract | CrossRef Full Text | Google Scholar

Radwan, M. A., AlQuadeib, B. T., Šiller, L., Wright, M. C., and Horrocks, B. (2017a). Oral administration of amphotericin B nanoparticles: antifungal activity, bioavailability and toxicity in rats. Drug Deliv. 24, 40–50. doi: 10.1080/10717544.2016.1228715

PubMed Abstract | CrossRef Full Text | Google Scholar

Radwan, S. E.-S., Sokar, M. S., Abdelmonsif, D. A., and El-Kamel, A. H. (2017b). Mucopenetrating nanoparticles for enhancement of oral bioavailability of furosemide: in vitro and in vivo evaluation/sub-acute toxicity study. Int. J. Pharm. 526, 366–379. doi: 10.1016/j.ijpharm.2017.04.072

PubMed Abstract | CrossRef Full Text | Google Scholar

Rana, V., and Sharma, R. (2019). “Recent advances in development of nano drug delivery,” in Applications of Targeted Nano Drugs and Delivery Systems, eds S.S. Mohapatra, S. Ranjan, N. Dasgupta, R.K. Mishra, and S. Thomas (Oxford: Elsevier), 93–131. doi: 10.1016/B978-0-12-814029-1.00005-3

CrossRef Full Text | Google Scholar

Salatin, S., Maleki Dizaj, S., and Yari Khosroushahi, A. (2015). Effect of the surface modification, size, and shape on cellular uptake of nanoparticles. Cell Biol. Int. 39, 881–890. doi: 10.1002/cbin.10459

PubMed Abstract | CrossRef Full Text | Google Scholar

Sarangapani, S., Patil, A., Ngeow, Y. K., Elsa Mohan, R., Asundi, A., and Lang, M. J. (2018). Chitosan nanoparticles' functionality as redox active drugs through cytotoxicity, radical scavenging and cellular behaviour. Integr. Biol. 10, 313–324. doi: 10.1039/C8IB00038G

PubMed Abstract | CrossRef Full Text | Google Scholar

Sayes, C. M., Aquino, G. V., and Hickey, A. J. (2016). Nanomaterial drug products: manufacturing and analytical perspectives. AAPS J. 19, 18–25. doi: 10.1208/s12248-016-0008-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Shan, X., Xu, T., Liu, Z., Hu, X., Zhang, Y.-D., and Wang, B. (2017). Safety and toxicology of the intravenous administration of Ang2-siRNA plasmid chitosan magnetic nanoparticles. Mol. Med. Rep. 15, 736–742. doi: 10.3892/mmr.2016.6090

CrossRef Full Text | Google Scholar

Sharifi, S., Behzadi, S., Laurent, S., Laird Forrest, M., Stroeve, P., and Mahmoudi, M. (2012). Toxicity of nanomaterials. Chem. Soc. Rev. 41, 2323–2343. doi: 10.1039/C1CS15188F

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, M., Sharma, S., Sharma, V., Sharma, K., Yadav, S. K., Dwivedi, P., et al. (2017). Oleanolic–bioenhancer coloaded chitosan modified nanocarriers attenuate breast cancer cells by multimode mechanism and preserve female fertility. Int. J. Biol. Macromol. 104, 1345–1358. doi: 10.1016/j.ijbiomac.2017.06.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, M., Shatkin, J. A., Cairns, C., Canady, R., and Clippinger, A. J. (2016). Framework to evaluate exposure relevance and data needs for risk assessment of nanomaterials using in vitro testing strategies. Risk Analysis 36, 1551–1563. doi: 10.1111/risa.12581

PubMed Abstract | CrossRef Full Text | Google Scholar

Shelma, R., and Sharma, C. P. (2011). Development of lauroyl sulfated chitosan for enhancing hemocompatibility of chitosan. Colloids and Surfaces B: Biointerfaces 84, 561–570. doi: 10.1016/j.colsurfb.2011.02.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, R. P., and Ramarao, P. (2013). Accumulated polymer degradation products as effector molecules in cytotoxicity of polymeric nanoparticles. Toxicol. Sci. 136, 131–143. doi: 10.1093/toxsci/kft179

PubMed Abstract | CrossRef Full Text | Google Scholar

Słoczynska, K., Powroznik, B., Pekala, E., and Waszkielewicz, A. M. (2014). Antimutagenic compounds and their possible mechanisms of action. J. Appl. Genet. 55, 273–285. doi: 10.1007/s13353-014-0198-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Sonaje, K., Lin, Y.-H., Juang, J.-H., Wey, S.-P., Chen, C.-T., and Sung, H.-W. (2009). In vivo evaluation of safety and efficacy of self-assembled nanoparticles for oral insulin delivery. Biomaterials 30, 2329–2339. doi: 10.1016/j.biomaterials.2008.12.066

PubMed Abstract | CrossRef Full Text | Google Scholar

Steimle, A., Autenrieth, I. B., and Frick, J.-S. (2016). Structure and function: lipid A modifications in commensals and pathogens. Int. J. Med. Microbiol. 306, 290–301. doi: 10.1016/j.ijmm.2016.03.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, J., Zhang, Q., Wang, Z., and Yan, B. (2013). Effects of nanotoxicity on female reproductivity and fetal development in animal models. Int. J. Mol. Sci. 14, 9319–9337. doi: 10.3390/ijms14059319

PubMed Abstract | CrossRef Full Text | Google Scholar

Tulinska, J., Kazimirova, A., Kuricova, M., Barancokova, M., Liskova, A., Neubauerova, E., et al. (2015). Immunotoxicity and genotoxicity testing of PLGA-PEO nanoparticles in human blood cell model. Nanotoxicology 9(Suppl. 1), 33–43. doi: 10.3109/17435390.2013.816798

PubMed Abstract | CrossRef Full Text | Google Scholar

VasanthaKumar, S., Ahamed, H. N., and Saha, R. N. (2014). Nanomedicine I: in vitro and in vivo evaluation of paclitaxel loaded poly-(ε-caprolactone), poly (dl-lactide-co-glycolide) and poly (dl-lactic acid) matrix nanoparticles in wistar rats. Eur. J. Drug Metab. Pharmacokinet. 40, 137–161. doi: 10.1007/s13318-014-0189-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Wallach, D., Kang, T.-B., and Kovalenko, A. (2013). Concepts of tissue injury and cell death in inflammation: a historical perspective. Nat. Rev. Immunol. 14, 51–59. doi: 10.1038/nri3561

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, H., Yu, X., Su, C., Shi, Y., and Zhao, L. (2018). Chitosan nanoparticles triggered the induction of ROS-mediated cytoprotective autophagy in cancer cells. Artif. Cells Nanomed. Biotechnol. 46(Suppl. 1.):293–301. doi: 10.1080/21691401.2017.1423494

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Q., Liu, P., Liu, P., Gong, T., Li, S., Duan, Y., et al. (2014). Preparation, blood coagulation and cell compatibility evaluation of chitosan-graft-polylactide copolymers. Biomed. Mater. 9:015007. doi: 10.1088/1748-6041/9/1/015007

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y., Zhou, J., Liu, L., Huang, C., Zhou, D., and Fu, L. (2016). Characterization and toxicology evaluation of chitosan nanoparticles on the embryonic development of zebrafish, Danio rerio. Carbohydr. Polym. 141, 204–210. doi: 10.1016/j.carbpol.2016.01.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Yah, C. S., Simate, G. S., and Iyuke, S. E. (2012). Nanoparticles toxicity and their routes of exposures. Pak. J. Pharm. Sci. 25, 477–491.

PubMed Abstract | Google Scholar

Yen, M.-T., Yang, J.-H., and Mau, J.-L. (2008). Antioxidant properties of chitosan from crab shells. Carbohydr. Polym. 74, 840–844. doi: 10.1016/j.carbpol.2008.05.003

CrossRef Full Text | Google Scholar

Yermak, I. M., Davidova, V. N., Gorbach, V. I., Luk'yanov, P. A., Solov'eva, T. F., Ulmer, A. J., et al. (2006). Forming and immunological properties of some lipopolysaccharide–chitosan complexes. Biochimie 88, 23–30. doi: 10.1016/j.biochi.2005.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Yildirimer, L., Thanh, N. T. K., Loizidou, M., and Seifalian, A. M. (2011). Toxicology and clinical potential of nanoparticles. Nano Today 6, 585–607. doi: 10.1016/j.nantod.2011.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Yostawonkul, J., Surassmo, S., Iempridee, T., Pimtong, W., Suktham, K., Sajomsang, W., et al. (2017). Surface modification of nanostructure lipid carrier (NLC) by oleoyl-quaternized-chitosan as a mucoadhesive nanocarrier. Colloids Surfaces B 149, 301–311. doi: 10.1016/j.colsurfb.2016.09.049

PubMed Abstract | CrossRef Full Text | Google Scholar

Yuan, Z.-Y., Hu, Y.-L., and Gao, J.-Q. (2015). Brain Localization and neurotoxicity evaluation of polysorbate 80-modified chitosan nanoparticles in rats. PLoS ONE 10:e0134722. doi: 10.1371/journal.pone.0134722

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, J., Tang, H., Liu, Z., and Chen, B. (2017). Effects of major parameters of nanoparticles on their physical and chemical properties and recent application of nanodrug delivery system in targeted chemotherapy. Int. J. Nanomedicine 12, 8483–8493. doi: 10.2147/IJN.S148359

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: hazard assessment, exposure assessment, in vivo toxicity, oxidative stress, genotoxicity, toxicity on reproduction, hemocompatibility, polymeric nanobiomaterials

Citation: Jesus S, Schmutz M, Som C, Borchard G, Wick P and Borges O (2019) Hazard Assessment of Polymeric Nanobiomaterials for Drug Delivery: What Can We Learn From Literature So Far. Front. Bioeng. Biotechnol. 7:261. doi: 10.3389/fbioe.2019.00261

Received: 02 August 2019; Accepted: 26 September 2019;
Published: 23 October 2019.

Edited by:

Michele Iafisco, Italian National Research Council (CNR), Italy

Reviewed by:

Smilja Markovic, Institute of Technical Sciences (SASA), Serbia
Ching-Yun Chen, National Health Research Institutes, Taiwan

Copyright © 2019 Jesus, Schmutz, Som, Borchard, Wick and Borges. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Olga Borges, olga@ci.uc.pt

Download