Next Article in Journal
Chitosan Nanoparticles Loaded with Capparis cartilaginea Decne Extract: Insights into Characterization and Antigenotoxicity In Vivo
Previous Article in Journal
Remote Loading: The Missing Piece for Achieving High Drug Payload and Rapid Release in Polymeric Microbubbles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Other Side of Plastics: Bioplastic-Based Nanoparticles for Drug Delivery Systems in the Brain

by
Erwin Pavel Lamparelli
1,†,
Marianna Marino
1,†,
Marta Anna Szychlinska
2,
Natalia Della Rocca
1,
Maria Camilla Ciardulli
1,
Pasqualina Scala
1,
Raffaella D'Auria
1,
Antonino Testa
3,
Andrea Viggiano
1,
Francesco Cappello
4,5,
Rosaria Meccariello
6,
Giovanna Della Porta
1,7,*,‡ and
Antonietta Santoro
1,7,‡
1
Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
2
Faculty of Medicine and Surgery, Kore University of Enna, Cittadella Universitaria, 94100 Enna, Italy
3
Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
4
Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
5
Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
6
Department of Movement and Wellbeing Sciences, Parthenope University of Naples, 80133 Naples, Italy
7
Research Centre for Biomaterials BIONAM, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work and share the last authorship.
Pharmaceutics 2023, 15(11), 2549; https://doi.org/10.3390/pharmaceutics15112549
Submission received: 20 September 2023 / Revised: 23 October 2023 / Accepted: 25 October 2023 / Published: 28 October 2023
(This article belongs to the Special Issue Nanocarriers for Drug Delivery in Tissue Engineering)

Abstract

:
Plastics have changed human lives, finding a broad range of applications from packaging to medical devices. However, plastics can degrade into microscopic forms known as micro- and nanoplastics, which have raised concerns about their accumulation in the environment but mainly about the potential risk to human health. Recently, biodegradable plastic materials have been introduced on the market. These polymers are biodegradable but also bioresorbable and, indeed, are fundamental tools for drug formulations, thanks to their transient ability to pass through biological barriers and concentrate in specific tissues. However, this “other side” of bioplastics raises concerns about their toxic potential, in the form of micro- and nanoparticles, due to easier and faster tissue accumulation, with unknown long-term biological effects. This review aims to provide an update on bioplastic-based particles by analyzing the advantages and drawbacks of their potential use as components of innovative formulations for brain diseases. However, a critical analysis of the literature indicates the need for further studies to assess the safety of bioplastic micro- and nanoparticles despite they appear as promising tools for several nanomedicine applications.

1. Introduction

Plastic diffusion is deemed a significant indicator of the onset of the Anthropocene, [1] an era in which humans altered and dominated the Earth and its ecosystems [2]. Despite the convenience aspects, the widespread use of plastics and their uncontrolled waste has resulted in negative impacts on the environment and human health [3]. In addition, most plastic products are added with various chemical compounds to improve functional properties, such as plasticizers (phthalates), flame retardants, antioxidants (i.e., IRGAFOS-168), acid scavengers, stabilizers (e.g., bisphenol A, BPA), and pigments [4]. Plastic degradation produces microplastics (MPs, particle size lower than 5 mm) and nanoplastics (NPs, size less than 1 µm) [5] that cross biological barriers and accumulate in the food chain [6].
More recently, “environmentally sustainable” plastic materials fabricated adopting biodegradable polymers, such as polylactic acid (PLA) and poly-lactic-co-glycolic acid (PLGA), have been introduced on the market. These bioplastics undergo a more rapid degradation in the environment; indeed, these biopolymers have been developed for pharmaceutical and biomedical applications in order to have a transient polymer (bioresorbable) but with mechanical properties similar to the non-biodegradable ones [7]. However, the recent widespread use of bioplastic-based materials needs a critical approach because the polymer’s faster biodegradation poses an issue for a more rapid accumulation in the form of particles in living tissues.
This issue is confirmed by the fact that these polymers are properly adopted in the form of micro- and nanoparticles as drug carriers in pharmaceutical formulations. For example, bioplastic-based NPs can be targeted and accumulated (depot systems) in a given tissue in order to achieve a proper sustained release of the loaded drug [8,9]. Bioplastic NPs can also prolong the therapeutic effect of a given drug, improving its efficacy [9]. Recent studies also suggest that specific concentrations of bioplastic NPs in the central nervous system (CNS) promote their passage through the blood–brain barrier (BBB) or blood–cerebrospinal fluid barrier (BCSFB). Indeed, bioplastic NPs seemed able to cross the BBB through transcytosis pathways and proper surface modifications can allow their passage through the BBB via receptor-mediated endocytosis or to deeply diffuse in the brain parenchyma [10]. This behavior, while extremely interesting for the development of new drug formulations for the CNS, poses significant challenges in terms of cost, failure, and clinical implementation; on the other hand, it may also raise public health concerns.
Hence, the aim of this review is to provide a different point of view on bioplastics and their degradation products. Truly, bioplastic MPs and NPs should be considered particularly effective for pharmaceutical formulations and precision medicine, to transport drugs into organs, like the brain, that are protected by biological barriers. Indeed, bioplastic MPs/NPs will be discussed as successful tools for brain drug delivery; however, different particle accumulation of bioplastics in the environment may enhance the issues for potential tissue accumulation, with unknown long-term biological effects.

2. Bioplastics: Definition, Chemical Properties, and Applications

Bioplastics can be divided into two categories: biodegradable and biobased [11,12,13]. Biobased plastics are entirely or partially made from biological resources and are not necessarily biodegradable. Plastics’ biodegradability is determined by the chemical composition of the polymer and environmental conditions [14]. On the other hand, biotic degradation is a process in which microorganisms, such as fungi or bacteria, reduce polymeric structure into smaller molecules that are utilized as a source of carbon or energy [15]. Photodegradation and hydrolysis consist of chemical processes in which high-energy radiations (UV) and water molecules induce polymer chain degradation [16,17,18]. In polymer degradation, biotic and abiotic factors can sometimes act together. Typically, abiotic degradation produces small fragments of plastic, which are subsequently degraded by microorganisms [18]. However, this process inevitably leads to the formation of small plastic particles with different characteristics and size [11,19].
Non-biodegradable plastics or petroleum-based plastics (conventional plastics) include polyethylene (PE), polypropylene (PP), polyvinyl chloride (PVC), and polystyrene (PS), which belong to the polyolefin class [20]. They are thermoplastic polymers in which olefin monomer units like ethylene, styrene, and vinyl chloride are combined to form long chains [21,22] (Figure 1). Polyolefins represent the leading industrial polymers due to their remarkable chemical stability and mechanical characteristics [23]. The manufacturing processes of these plastics and applications have been well described elsewhere [24,25,26,27,28,29,30,31,32,33,34]. The so-called “biobased” plastics, such as bio-PE obtained from sugar cane [35] and bio-PET produced by the oxidization of bio-ethylene derived from the fermentation of glucose [36], find many applications in the packaging sector, particularly for drinking bottles and textile industries; however, they are non-biodegradable [37].
Biodegradable plastics comprise poly-caprolactone (PCL), poly-butylene succinate (PBS), poly-butylene adipate terephthalate (PBAT), poly-lactic acid (PLA), poly-lactic-co-glycolic acid (PLGA), and poly-hydroxy-alkanoate (PHA) [14] (Figure 1). PCL is suitable for a wide range of medical applications such as implantable biomedical devices, sutures, and tissue engineering scaffolds due to its biocompatibility and slow degradation [38,39,40]. It is used for the synthesis of green polyurethane [41] and commonly blended with biobased biodegradable plastics [42] to improve its thermal and mechanical properties [43]. On the contrary, PBS has a relatively slow biodegradation rate and biocompatibility [43].
PLA is made from 100% bioresources and is totally biodegradable and recyclable [44]. It is produced by a combination of lactic acid monomers derived from the fermentation of sugars obtained by sugar cane, potatoes, and corn [45,46]. PLA degrades in 6 to 24 months in the environment, depending on various factors, such as temperature, product size and shape, and isomer ratio. Despite some inherent weaknesses like brittleness and moisture uptake, it exhibits good thermomechanical properties like the traditional plastics PET and PP and has been extensively applied in different fields, ranging from packaging applications, bowls, films, and bottles, to clothes, textile furniture, hygiene products, and mulch films for agriculture [47,48,49]. Furthermore, it can be also copolymerized with polyethylene glycol (PEG) to enhance its hydrophilic and biocompatibility properties, making it suitable for drug delivery systems [50]. However, despite its eco-friendly characteristics, the commercial production of PLA is hindered by the high cost of raw materials and the lack of composting infrastructure in most markets. Implementing composting infrastructure would enable the widespread use of PLA and would reduce the environmental impact of traditional plastics [51].
PLGA is another bioplastic component frequently used as a copolymer of polyglycolic acid (PGA) and PLA. In fact, it is frequently employed in biomedical applications, due to its biocompatibility and fast biodegradation. PLGA, like PLA, can be produced by polycondensation or ring-opening polymerization, varying its molecular weights and monomer ratios to ameliorate its degradation rate [52].
PHAs are aliphatic polyesters synthesized through the polymerization of b-, g-, and d-hydroxyalkanoic acids obtained from the fermentation of sugars and lipids from various feedstocks [53]. PHAs are polymerized by bacteria, which can synthesize them under stressful conditions as a carbon and energy reserve [54]. Large-scale production of PHAs is expensive, requiring fermentation, isolation, and purification processes that limit their widespread use [55,56]. Nonetheless, PHAs are driving the growth of the biodegradable bioplastics market, with production capacity expected to triple in the next five years [57]. Poly-4-hydroxybutyrate (PHB) and poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) are the most commonly used PHAs: PHB has a high elastic modulus and better barrier properties than PLA. However, it is brittle and has lower thermal stability. PHBV has PP-like properties and is commercially available added with a hydroxyvalerate (HV) that is able to confer more flexibility than PHB [58]. Like PLA, PHAs find various applications in the packaging and biomedical fields as single-use items. PHB and PHBV have been investigated for their potential use as bioresorbable materials for surgical sutures, wound dressings, tissue scaffolds, bone fracture fixation plates, and porous sheets for tissue regeneration in injured soft tissues [53]. The biodegradability of PHAs depends on factors, such as chain configuration, crystallinity, and processing conditions. Another important advantage of PHAs is their high degradation rate in marine environments [59].

3. Routes for MPs and NPs Adsorption, Tissue Accumulation, and Biological Effects

All plastics, both biodegradable or not, can initiate their degradation process reducing the manufacture size into smaller particles known as MPs and NPs. Studies on preferential adsorbing routes, tissue internalization, accumulation, and molecular mechanisms of penetration are important to assess toxic potential and to better understand the capability of MPs and NPs to target specific tissues. Indeed, there is growing evidence of the harmful effects of MPs/NPs on organisms ranging from plants and fish to microorganisms and animals [60]; thus, MPs/NPs have been studied for their potential hazards and health implications for humans even though investigations are still in infancy.

3.1. Dermal Route

Due to the presence of bioplastic MPs/NPs in cosmetic products, such as body and facial scrubs, creams, soaps, and other beauty products, as well as in microfibers and drug delivery systems for dermal application, the skin represents a potential route for human exposure [61]. For instance, hair follicles, sweat glands, and injured skin represent all possible entry routes [61]. Some interesting data on conventional NPs’ dermal penetration were obtained by Campbell et al. [62], who demonstrated that PS-NPs reach a depth of approximately 2–3 µm from the top layers of pig skin tissue. Furthermore, Vogt et al. [63] demonstrated the presence of 40 nm-diameter fluorescent PS-NPs in the perifollicular tissue of human skin explants undergone cyanoacrylate follicular stripping, indicating a size-dependent absorption of transcutaneous application of particles by Langerhans cells around hair follicles. By using ex vivo human skin samples, Zou et al. [64] investigated the effects of skin condition, incubation temperature, particle size, and vehicle solutions on NPs’ uptake. They found that the use of dimethyl sulfoxide as a vehicle led to deeper penetration of PS-NPs compared to ethanol and water. Tape stripping also allowed deeper penetration, but only until the granulosum layer. Similarly, in excised human skin samples, Jatana et al. [65] observed that ingredients present in skincare lotions can facilitate bioplastic particle penetration of PCL-NPs corroborating that skin conditions and vehicles influence the uptake of these NPs by the skin. Considering animal and human skin differences in their anatomical structure, results obtained by ex vivo human samples should be considered more reliable [66]. The overall observations suggest that the dermal penetration route is not preferred; thus, bioplastic MPs/NPs are not largely used in pharmaceutical applications for transdermal drug delivery. However, the effects of this route on human health received very little attention and should be certainly further investigated.

3.2. Inhalation

The fact that MPs and NPs can be transported in the air over long distances favoring their presence both in aquatic and terrestrial environments has been well documented in conventional plastics, whereas studies on bioplastics are not [67,68,69]. Due to their size, MPs/NPs can be easily inhaled and can reach the lungs, where they should be removed through the mucociliary escalator or phagocytosis by alveolar macrophages [70], but MPs/NPs can overcome these defense mechanisms and remain on the alveolar surface, causing lung damage. In vitro studies report a positive correlation between exposure to MPs and the development of pulmonary inflammation and cancer [70,71]. Exposure to MPs (4.06 ± 0.44 μm) to human lung epithelial cells (BEAS-2B) has been shown to induce the formation of reactive oxygen species (ROS), inflammatory process, and cytotoxic effects, as well as a decrease in transepithelial electrical resistance [72]. The onset of these harmful processes is affected by MPs/NPs’ properties, such as hydrophobicity, surface charge, and functionalization, that influence their absorption and clearance in the lungs. Among these properties, particle size rather than particle kind has a great relevance: the smaller the particles, the greater the effect and distribution (see Table 1). For example, in rat models, the instillation of ultrafine PS-NPs of different sizes (from 64 to 535 nm) caused lung inflammation. This effect was mediated by an excessive neutrophil influx and increased pro-inflammatory proteins and lactate dehydrogenase in bronchoalveolar lavage (BAL) [73]. Accordingly, in alveolar cell lines (A549), the treatment with PS-MPs determined an increase in IL-8 mRNA expression after 2–4 h of treatment [73]. In addition, it has been found that a mixture of MPs (MPs < 50 μm) taken from environmental plastic waste can accumulate in A549 cells exposed in vitro and induce oxidative stress, genotoxicity, and alterations in gene expression [74,75]. Similar results were obtained by exposing mice to particles from tire wear by inhalation [76]; in this case, reduced ventilatory functions and exacerbated pulmonary fibrotic injury were observed [76].
The alveolar epithelium barrier is thin enough for NPs to enter blood capillaries, allowing them to disperse throughout the human body and potentially cross the BBB [77]. Both conventional and bioplastic particles, depending on their hydrophilicity, size, and surface charge, can translocate and enter the circulation, probably more easily when endothelial and epithelial permeability is increased during immune/inflammation response. In fact, a research group aiming to develop improved mucosal immunization strategies exposed BALB/c mice to PS carboxylate MPs (1.1 µm in size) by intranasal delivery. Treated mice exhibited the presence of PS-MPs in the nasal-associated lymphoid tissues (NALTs) and draining cervical lymph nodes after 7 days. In addition, MPs’ accumulation in the spleen was also observed corroborating that the spleen can act as an inductive site following bronchopulmonary deposition [78].
Since the nasal route is also used for drug delivery to the brain [79], it can be argued that once MPs/NPs are unintentionally inhaled, the olfactory nerve might permit the transit of NPs to the CNS. Different studies have demonstrated that various nanomaterials, once in contact with the olfactory epithelium, can be transported to the brain through olfactory neurons inducing detrimental effects such as brain inflammation [80,81,82]. However, few studies have investigated this exposure route, even if some authors confirmed the presence of MPs and NPs in the respiratory system and their passage through the BBB [67,70]. Collectively, both in vitro and in vivo research suggest that MPs/NPs have toxic effects on the respiratory tract and lungs, leading to inflammation and lung fibrosis.
Table 1. Preferential accumulation sites of plastic or bioplastic MPs/NPs’ exposure in vivo and their biological effects.
Table 1. Preferential accumulation sites of plastic or bioplastic MPs/NPs’ exposure in vivo and their biological effects.
Exposure RouteTypeSizeAccumulationBiological EffectsRef.
Dermal contactNPs20–200 nmHair follicular openingsND[83]
NPs40 nm
750 nm
1500 nm
Langerhans cells and epidermal cellsND[63]
InhalationNPs64 nmLung epitheliumLung inflammation, excessive neutrophil influx, proinflammatory proteins[73]
NPs<1 μmPulmonary alveolar unitsPulmonary parenchymal lesion, alveolar stenosis, fibrous tissue hyperplasia, perivascular, lymphocyte, infiltration,
ꜜ E-cadherin expression,
ꜛ collagen deposition
[76]
MPs1.1 μmNALTs, mediastinal lymph node, spleen, bronchopulmonary depositionꜛ Immunological response[84]
IngestionMPs10–150 μmColon and duodenumGut microbiome alterations, intestinal inflammation,
ꜛ pro-inflammatory cytokines, intestinal glands disruptions
[85]
NPs100 nmStomach, small and large intestines, kidney, lungs Liver immune cells infiltration, hepatocyte vacuolization, pulmonary interstitial fibrosis, renal tubular atrophy, ileum epithelium disruption, colon lymphocyte aggregation, neuron alterations, testicular atrophy[86]
MPs/NPs50 nm
500 nm
5 μm
Intestine, liver, kidney, testis, brainꜛ Inflammatory factors[87]
Abbreviations: MPs, microparticles; NPs, nanoparticles; PS, polystyrene; TW, tire wear; PE, polyethylene; NALTs, nasal-associated lymphoid tissues. ꜛ increase; ꜜ decrease.

3.3. Ingestion

The most significant MPs/NPs exposure route for humans is ingestion. MPs/NPs are present in food and drink containers, and in edible products, and have been found in the gastrointestinal system of fishes [5,84,88]. As in skin and lungs, in gastric cells, smaller-sized particles have more possibilities to be absorbed. Many studies are available in the current literature examining the effects of conventional plastics and their accumulation in the gastrointestinal tract, while data on the consequences of the ingestion of bioplastics are still lacking. In a study carried out by Banerjee et al. [89], the toxicity of PS MPs/NPs was examined in SNU-1 human gastric epithelial cells. The study revealed that smaller particles (50 nm) were taken up more avidly by the cells than larger particles (1000 nm), increasing cell cytotoxicity, apoptosis, and necrosis [89]. To gain a better understanding of the chronic exposure to MPs/NPs through the gastrointestinal tract, Domenech et al. (2021) [90] investigated the effects of 50 nm PS particles on CaCo-2 colon cancer cells. After 8 weeks of treatment, it was found that 20% of the cells had taken up and internalized the particles and exhibited altered expression of oxidative-stress-related genes. Conversely, another study [85] demonstrated that 5 weeks of exposure of mice to PE-MPs by ingestion led to gut microbiome alteration and intestinal inflammation. The MPs/NPs-induced effects were not limited to the gastrointestinal tract since the potential ability of NPs to permeate the gut epithelium and pass into the systemic circulation can lead to both damage and disruption of the intestinal barrier and accumulation in other tissues and organs far from the primary route of exposure [91]. Accordingly, Xu et al. [86] investigated the uptake mechanism of NPs in mice demonstrating that PS-NPs’ absorption occurs through clathrin-mediated endocytosis. Furthermore, a reduction in occludin and zonula occludens-1 (ZO-1) was observed in Caco-2 cells, thus suggesting the potential ability of these particles to disrupt the intestinal barrier. By passing the intestinal epithelium, particles could enter the circulation accumulating in the spleen, liver, lung, and brain [86]. Han et al. [87] demonstrated that PS particles combined with di-(2-ethylhexyl) phthalate (DEHP) accumulated in mouse organs, such as intestines, livers, kidneys, testis, and brain. The accumulation of NPs in the brain after ingestion probably takes place through the lymphatic or blood circulation, the translocation through the BBB—which appears to be affected by enhanced permeability induced by PS-MPs—and the transport up to the brain parenchyma [21,92,93].
Noteworthy, after entering the circulatory system, MPs and/or NPs could also affect blood and immune cells. Indeed, we have shown that PLA microbeads (size 1 ± 0.2 μm) used as drug delivery systems can be internalized by human monocytes with an efficiency of 30%; but after PLA-MPs phagocytosis, cell apoptosis increased in a dose-dependent manner [94]. Instead, empty microcarriers of PLGA (mean size 827 ± 68 μm) at a concentration of 12.5 mg/mL did not induce any cytotoxic effect on human peripheral mononuclear cells (hPBMCs) from healthy donors [95]. By reducing the size of PLA- and PLGA-NPs (0.4–3 μm), both PLA and PLGA NPs exhibited low cytotoxicity in Chinese hamster ovary (CHO) cells and in hPBMCs, corroborating, as in conventional NPs, that the particle size is fundamental to cause biological effects. However, at the same concentration, PLGA affected cell viability more than PLA [96].
Studies on contamination routes, deposition, and related health risks of exposure to bioplastic MPs/NPs are widely unexplored. Few data indicated that gut enzymatic hydrolysis of PLA-MPs in mice generated smaller particles by competing for triglyceride-degrading lipase. PLA oligomers and their NPs accumulate in the liver, intestine, and brain, inducing intestinal damage and acute inflammation [97]. Details regarding the size and distribution of MPs/NPs in tissues are given in Table 1, while a schematic representation showing different aspects of adsorption, tissue accumulation, and biological effects is summarized in Figure 2.

4. Bioplastic-Based Polymeric NPs for Drug Delivery in the Brain

4.1. Bioplastic MPs/NPs as Drug Delivery Systems

Significant advancements in nanomedicine have led to the development of drug delivery systems based on bioplastic MPs/NPs. These systems offer several advantages, including increased drug shelf-life, targeting to the specific organ/tissue, reduced therapeutic dose administrations, improving drug effectiveness, and minimizing side effects. Among drug delivery systems, the PLA and PLGA-NPs are the most extensively studied [98], because of their high drug-loading capacity, good biocompatibility, biodegradability, and tunable release properties. PLA/PLGA pharmaceutical formulations were developed for sustained delivery of several drugs used in cancer treatment, anti-inflammatory compounds, and drugs for neurological disease [99,100], while many others are still in phase II or III of clinical trials [101] (see Table 2).
In the field of treatment of neurodegenerative diseases, pharmaceutical formulations have been often unsuccessful due to the BBB that is very difficult to cross [102]. Instead, PLA/PLGA NPs have emerged as a promising solution for delivering drugs to the brain, as they can overcome the physiological barrier through different strategies, such as transcytosis pathways (Trojan horse strategy) or by escaping the efflux pumps by bearing specific ligands onto the particle surface [10]. In general, the smaller size coupled with surface modifications improved formulation pharmacokinetics, enhancing cell uptake and, consequently, drug absorption [103,104].
Table 2. Bioplastic MPs/NPs-based drug delivery systems approved by the FDA for disease treatments.
Table 2. Bioplastic MPs/NPs-based drug delivery systems approved by the FDA for disease treatments.
NameBioplasticLoaded DrugTherapeutic ApplicationCompanyFDA Approval (Date)Ref.
Lupron Depot® PLGA Leuprolide acetateProstate cancer, endometriosisTakeda–Abbott Products (Osaka, Japan)1989[105]
Atridox®PLADoxycycline hyclateChronic adult periodontitisTolmar (Fort Collins, CO, USA)1998[99]
Sandostatin Lar® PLGAOctreotide acetateAcromegalyNovartis (Mulgrave, VIC, Australia)1998[99]
Trelstar®PLGATriptoreline pamoateAdvanced prostate cancerAllergan
(Gordon, NSW, Australia)
2001[99]
Risperdal Consta® PLGARisperidoneSchizophrenia, bipolar I disorderJanssen (Beerse, Belgium)2003[106]
Vivitrol® PLGANaltrexoneAlcohol dependenceAlkermes (Waltham, MA, USA)2006[100]
Signifor Lar® PLGAPasireotide pamoateAcromegalyNovartis (Mulgrave, VIC, Australia)2014[99]
Sublocade® PLGABuprenorphineOpioid disorderIndivior (Richmond, VA, USA)2017[99]
Triptodur Kit® PLGATriptorelin pamoateCentral precocious pubertyArbor (Mulgrave, VIC, Australia)2017[99]
Scenesse®PLGAAfamelanotidePrevention of phototoxicity in erythropoietic protoporphyriaClinuvel (Melbourne, VIC, Australia)2019[107]
Durysta®PLA/PLGABimatoprostGlaucoma, open-angle, intraocular hypertensionAllergan (Gordon, NSW, Australia)2020[107]
Abbreviations: PLGA, polylactic co-glycolic acid; PLA, polylactic acid; PCL, polycaprolactone.

4.2. Techniques for Fabricating Bioplastic MPs/NPs

Several technologies have been described for the fabrication of bioplastic MPs/NPs, such as nanoprecipitation, solvent evaporation or extraction, spray drying, and supercritical fluids [108,109]. All described technologies are schematically illustrated in Figure 3, and examples of MPs/NPs’ characterizations by laser scattering (size distribution curve) or SEM/TEM (micrographs) are shown in Figure 4.
The nanoprecipitation process involves the precipitation of a polymer from a water-miscible organic solvent by mixing it with an aqueous medium, acting as an anti-solvent (see Figure 3). The size can be controlled by adjusting different parameters, such as organic solvent, polymer concentration, and surfactant amount in the water [110,111,112]. Since proper mixing is required between the two fluids, microfluidic systems have been recently described as promising tools for this task [113]. Indeed, the nanoprecipitation within micrometering channels assures strict control over the particle size by varying pump flow rates and micromixer geometry [114,115]; however, the method is unsuitable for encapsulating hydrophilic drugs into the NPs [112] (Figure 3b).
Solvent evaporation from emulsion is also widely used; that is, when emulsions undergo evaporation or extraction, the dispersed oily droplets within the surrounding water phase can solidify due to organic solvent removal, developing micro/nanocarriers (Figure 3c) [116,117,118]. Physical properties of resulting particles can be modulated by varying surfactant type and concentration, stirring rate, and solvent evaporation conditions; the method can encapsulate hydrophobic and hydrophilic drugs depending on the use of single or multiple emulsions; however, the evaporation step demonstrates fluctuations in reproducibility from one batch to another [119,120]. At the same time, extraction requires comparatively large amounts of a second solvent, with the related issue of further solvent recovery. Both processes require processing times of several hours that can promote aggregation phenomena between the droplets, producing carriers with a larger polydispersity [121]. It should also be noted that, despite the widespread use of solvent evaporation/extraction processes to prepare polymeric carriers, there are no established standard protocols, and each preparation follows its own set of procedures. Finally, this may not be suitable for temperature-sensitive drugs due to the risk of degradation during the solvent evaporation step as well as poor encapsulation efficiency of high water-soluble compounds reported [95].
Dense carbon dioxide technologies have been proposed to produce bioplastic MPs/NPs for different drug delivery purposes [122,123,124,125] or tissue engineering [96,126]. Supercritical emulsion extraction (SEE) technology operating in a continuous layout using a counter-current packed tower [127,128] was described for both PLA and PLGA carriers’ fabrication. In detail, an emulsion that contains polymer undergoes solvent removal using supercritical carbon dioxide as an extraction fluid (Figure 3d). The dense-gas extraction technology ensured improved performances, such as a better batch-to-batch reproducibility [129], more accurate carrier size control—thanks to a fixed droplet shrinkage without aggregation phenomena [130], lower solvent residue, and better-controlled encapsulation efficiency [95,96,131].
Nano spray drying is a relatively recent technique enabling the single-step fabrication of bioplastic MPs/NPs starting from a small sample volume. In this process, a liquid solution containing both the polymer and the drug is transformed into tiny droplets through atomization by a spray nozzle. Subsequently, these droplets undergo rapid drying as they are exposed to a stream of hot air or inert gas. As a result, solid particles are formed through the evaporation of the solvent (Figure 3e). In this case, the particle size and especially the size distribution may be impacted by electrospray process parameters, such as nozzle diameter, spraying rate, and drying temperature [132]. This method can yield nanoparticles with a narrow size distribution and high drug-loading capacity [133]. In contrast, conventional spray drying can produce carriers with lower encapsulation efficiency and larger size and granulometry.
To sum up, the choice of suitable technology for the production of bioplastic MPs/NPs for drug delivery will depend on several factors, including polymer or drug solubility, desired carrier size, distribution, and shape or surface charge.

4.3. Bioplastic MPs/NPs Delivery to the Brain

In general, a drug delivery system facilitates the attainment of the desired therapeutic response of an active substance by enhancing its bioavailability at the target site while ensuring optimal effectiveness and safety [134]. Due to the presence of the BBB, which limits the entrance of external substances into the brain, many efforts are being made to use bioplastic NPs as drug carriers to the brain. Although parenteral administration is the prevalent route for bioplastic MPs/NPs, its effectiveness for brain drug delivery is still in development. Intranasal administration is an alternative route to bypass the BBB as it allows direct access to the brain, even though its clinical application is hindered by the limited knowledge of nanoparticle deposition and absorption in this anatomical site [135]. Intracranial administration, by bioplastic MPs/NPs’ injection directly into the brain tissue or cerebrospinal fluid, is a direct and effective route for brain drug delivery using bioplastic MPs/NPs. However, this route is invasive and may cause tissue damage or inflammation [136]. Intrathecal administration is another direct route for brain drug delivery that involves the injection of bioplastic MPs/NPs into the spinal cord or cerebrospinal fluid [137]. This route accounts for the targeted delivery of NPs to the brain and has shown to be the most promising for brain tumors and neurodegenerative disease treatment [137]. Overall, the intravenous route remains the preferred choice as those mentioned before are too invasive. As a result, several strategies have been devised to overcome the BBB and improve the delivery of bioplastic MPs/NPs to the brain. One strategy is to modify the surface of bioplastic MPs/NPs with BBB-penetrating molecules, such as peptides, antibodies, or aptamers. These modifications can increase the affinity of NPs to the BBB and enhance their transport across the barrier [138]. Another strategy involves employing ultrasound or magnetic fields to increase BBB permeability and enhance the transport of bioplastic MPs/NPs’ carriers [139].
Recently bioplastic MPs/NPs have been engineered to overcome the BBB or target specific cell types in the brain, such as neurons or glial cells. These targeted nanoparticles can enhance drug delivery to specific regions of the brain and reduce off-target effects [140]. Essentially, there are three distinct strategies to accomplish this objective: adsorptive-mediated transcytosis, transporter-mediated transcytosis, and receptor-mediated transcytosis. The first mechanism can be facilitated through electrostatic interactions between the negatively charged components present on the luminal surface of cerebral endothelial cells and the cationic groups or conjugating specific compounds, such as lectins, cardiolipin, and heparin to the NPs surface [141]. Another approach for delivering drugs to the brain is utilizing transporter-mediated transcytosis. Indeed, it is feasible to synthesize nanoparticles with surface-conjugated molecules (glucose and its analogs, glutathione, and amino acids) that exhibit strong recognition by the transporters that are overexpressed in brain endothelial cells [142]. The last approach to access brain tissue involves the use of receptors overexpressed within the BBB. In more detail, transferrin, lactoferrin, low-density lipoproteins, and nicotinic acetylcholine receptors are commonly employed receptors to achieve receptor-mediated transcytosis across the BBB [140].
Finally, the use of prodrug strategies can also enhance the delivery of bioplastic MPs/NPs to the brain. Prodrugs are inactive precursors of active drugs that can be activated within the brain by specific enzymes. In fact, hydrophilic and high-molecular-weight compounds cannot traverse the BBB or the blood–cerebrospinal fluid barrier (BCSFB) through paracellular pathways. In contrast, lipophilic solutes can passively permeate these barriers [143]; hence, hydrophilic drugs can be chemically modified into lipophilic prodrugs by concealing polar functional groups. This approach can increase the brain concentration of active drugs while reducing their systemic toxicity [144].
Altogether, the described strategies offer promising solutions to overcome the BBB and enhance the delivery of bioplastic NPs to the brain.

5. Biological Effects of Bioplastic NPs Loaded Carriers in the Brain

5.1. Bioplastic MPs/NPs for Brain Diseases: Advantages

Several PLGA-NPs have been developed for Alzheimer’s disease (AD) treatment. Yusuf et al. [145] designed PLGA-NPs loaded with the phytochemical compound thymoquinone (TQ). The TQ-loaded PLGA-NPs, coated with polysorbate 80, successfully crossed the BBB and significantly increased superoxide dismutase (SOD) activity in male albino mice [145,146]. Furthermore, Xu et al. [147] synthesized Tween 80-coated methoxy poly (ethylene glycol) PLGA-NPs loaded with rhynchophylline (RIN), a spirocyclic alkaloid with neuroprotective effects, to target the brain for AD treatment. In vitro and in vivo studies demonstrated that both these bioplastic NPs had a high transport across BBB and improved survival rate of neuronal cells [148]. Since AD is linked to Aβ aggregation, PEGylated bioplastic NPs loaded with an Aβ1–42 antibody have been developed and its efficacy has been investigated in a transgenic AD mouse model [149]. The treatment led to a significant improvement in memory and to a reduction in Aβ levels in the brain, indicating the potential of this approach for treating AD.
Even for the treatment of Parkinson’s disease (PD), several PLGA-NP systems have been developed. This neurodegenerative disorder is characterized by tremors, dyskinesia, and motor impairments. The appearance of these symptoms is attributable to a degeneration of dopaminergic neurons within the substantia nigra with consequent destruction of the nigrostriatal pathway [150,151,152]. Current therapies for PD management aim to enhance dopamine levels; in fact, the gold standard for the treatment of PD is the administration of L-Dopa, a dopamine precursor [153]. In line with such strategies, Monge-Fuentes et al. [154] developed bioplastic NPs loaded with dopamine and composed of albumin and PLGA. This nano-system, thanks to albumin, can cross the BBB better than free L-DOPA. PD mice, administered with these L-DOPA-loaded bioplastic NPs, show augmented levels of dopamine and improvements in motor symptoms [154]. Accordingly, PLGA-NPs, conjugated with wheat germ agglutinin (that enhances absorption via the nasal cavity) and loaded with L-Dopa, showed a significant improvement in the drug delivery to the brain together with better therapeutic efficacy and lower side effects [155].
More complex bioplastic NP systems have been developed consisting of multilayer hybrid PLCL-NPs encapsulating together L-Dopa, Tenoxicam as an anti-inflammatory drug, and Lamotrigine as a neuroprotective agent. This formulation was able to act on more than one complication associated with PD and improved cognitive abilities in PD-induced rats [156].
Few PLGA-based drug delivery systems have been developed for the treatment of other neurological disorders, such as Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). HD is a genetic disease characterized by an accumulation of the mutant huntingtin (mHTT) protein in nerve cells [157]. Symptoms consist of motor impairment, psychiatric disorders, and behavioral disorders as a consequence of the progressive loss of nerve cells and brain mass; the neurons of the striatal part of the basal ganglia are the most affected [157]. Currently, there are no approved therapies to delay the onset and progression of HD. Therapeutic approaches targeting the cause of HD, i.e., the CAG-expanded HTT gene and its products, or downstream processes associated with the pathogenesis of HD, are still in the clinical development [158]. Joshi et al. [159] designed PLGA-NPs coated with polysorbate 80 and loaded with oligonucleotides able to inhibit the HTT gene. These authors, using a Drosophila model of HD, reported improvements in motor performance without signs of toxicity [159,160].
For the treatment of ALS, a neuromuscular disease in which motor neuron loss occurs due to an altered expression of retinoid signaling [161,162] realized adapalene-loaded PLGA-NPs. These bioplastic NPs, tested in OD1G93A transgenic mice, showed an activation of the retinoid signaling pathway in the CNS with an improvement in neuroprotection and motor performance.
MS is known as a disease in which the immune system attacks the myelin sheath, an important component of axonal membranes, inducing progressive loss of neuronal structure and functions [163]. In this regard, recently Gholamzad et al. developed PLGA-NPs conjugated with myelin oligodendrocyte glycoprotein (PLGA-MOG) [164]. These particles were then intravenously administered to a C57BL6 mouse model of MS. PLGA-MOG NPs were able to ameliorate clinical symptoms and autoimmune responses, reducing the infiltration of immune cells within the brain.
Bioplastic NPs have emerged also as a strategy for the treatment of brain cancers such as glioblastoma which is considered the most aggressive form of CNS tumors [165,166]. In a study carried out by Maksimenko et al. [167], it emerged that PLGA-NPs (size 110 nm), loaded with doxorubicin and coated with poloxamer 188 (Dox-PLGA), crossed the BBB more efficiently than doxorubicin alone in a xenograft rat model. Dox-PLGA-NPs were able to significantly reduce intracranial 101.8 glioblastoma size after 2 weeks of treatment with 3 × 1.5 mg/kg bw (as doxorubicin) after tumor implantation. Accordingly, some other studies carried out to improve the transport of anti-proliferative drugs across the BBB have shown that transferrin-conjugated magnetic silica PLGA-NPs loaded with doxorubicin and paclitaxel stimulate ROS and TNF-α production inducing time and dose-dependent cytotoxicity against glioma cells in vitro and in vivo [168]. The effectiveness of conjugated nanocarriers loaded with chemotherapeutic drugs has also been demonstrated for PLGA-NPs—loaded with morusin (PLGA–MOR) and conjugated with chlorotoxin (CTX), a peptide that binds specific chloride channels and matrix metalloproteinase (MMP-2) [169]. In this case, the treatment of U87 and GI-1 glioma cells with PLGA–MOR–CTX NPs resulted in enhanced anti-proliferative effects by inhibiting MMP activity and inducing cytoskeletal alterations, ROS generation, and apoptosis [169].
The overall data indicate undoubtedly that bioplastic NPs represent a promising strategy for the treatment of neurodegenerative disorders and brain tumors, since they can cross or bypass the BBB delivering drugs in a specific manner and preserving their efficacy. Despite the numerous efforts and encouraging results obtained until now, we are still far from the effective use of these formulations, as no bioplastic NPs-based system is currently on the market [170]. The only available data concern preclinical studies (Table 3), and many aspects mainly linked to pharmacokinetics (ADME) still need to be clarified [171,172].

5.2. Bioplastic MPs/NPs for Brain Diseases: Drawbacks

Despite bioplastic MPs/NPs seeming effective as drug delivery systems for the brain, their toxicity is largely unknown, and available information is strictly related to conventional NPs from industrial production. In this regard, several studies have demonstrated that NPs could be involved in neuroinflammation and increase oxidative stress [172,174,175]. The generation of ROS could cause damage to lipids, nucleic acids, proteins, and other essential biomolecules, leading to mitochondrial dysfunction and cell death by altering signaling pathways [176]. Bioplastic MPs/NPs’ exposure is also responsible for activation of microglia cells and astrocytosis, leading to neuroinflammation and neuron function impairments [177]. However, to date, toxicological data are still few and limited to cautious hypotheses referring to systemic toxicity. In addition, the only available data differ from each other by the administration route, dose metrics, particle size, and loaded drug [178], making it difficult to draw general conclusions. Indeed, despite PLGA and PLA biocompatibility and biodegradability, bioplastic MPs/NPs for drug delivery, when produced on a nanoscale dimension, could have toxic effects [179] that are far from being fully understood. Bioplastic MPs/NPs may have dimensions quite far from cells (1:10; 1:100), and this implies that they could easily interfere with the activity and metabolism of neurons, microglia, and astrocytes [180,181]. A study focused on the in vivo neurotoxicity of polysorbate 80-chitosan NPs, after intravenous injection in rats, showed an accumulation in several brain areas such as the frontal cortex and cerebellum inducing neuronal death, inflammation, and oxidative stress [182]. Furthermore, the removal of bioplastic MPs/NPs by the blood can occur very slowly, thus promoting their accumulation with potential side effects [183]. From this point of view, it is crucial to analyze damaging effects at different levels, including the ultrastructure of the cell, since this level represents the boundary between molecular and spatially organized levels of the body. NPs interact with the cell precisely at this level, and dysfunction of cells or the action of redundant and regulatory systems could play a negative role in smoothing out the effectiveness of treatments. Another important factor is the toxicity related to degradation. PLA or PLGA degradation products, such as lactic or glycolic acid, could lead to tissue acidification when accumulated. Furthermore, PLGA has been reported to be much more responsible for inflammation because of its faster degradations (weeks) with respect to PLA (months) [184]. Increased levels of lactate could interfere with bioenergetic processes such as glycolysis and oxidative phosphorylation, and it is known that brain alterations in energy metabolism are associated with schizophrenia and bipolar disorders [185]. Furthermore, bioplastic MPs/NPs once in contact with biological systems could interact with neighbor biomolecules. As a result, these interactions could lead to the formation of the so-called “protein corona”. Protein corona affects NPs’ properties and alters their pharmacokinetics with possible aberrating biodistribution, toxicity, and mistargeting [186,187,188].
Other several factors could influence the neurotoxicity of bioplastic MPs/NPs. First of all, the size of polymeric NPs: all NPs with smaller sizes can penetrate the BBB more easily and have a higher surface area which can increase their interactions with brain cells and cause toxic effects [189]. The surface chemistry of bioplastic MPs/NPs is another factor that can influence their neurotoxicity. Surface modifications such as the addition of targeting ligands or PEGylation could alter the surface charge and hydrophobicity of NPs, affecting their cellular uptake but also their ability to interfere with endogenous cell signaling pathways [140]. Furthermore, intravenous injection can cause the accumulation of bioplastic MPs/NPs in the liver, spleen, and other organs, leading to potential systemic adverse effects. Finally, higher doses and more frequent administrations can increase the negative health effects, while lower doses and infrequent administration can reduce the toxic risk [190]. In summary, bioplastic MPs/NPs possess good properties for successful use in pharmaceutical preparations for the treatment of brain diseases; but it is important to understand the balance between the therapeutic and toxic actions of these drug carriers, because their potential clinical use still represents a matter of concern (Figure 5).

6. Conclusions

In the light of environmentally friendly solutions, nowadays bioplastics are considered a valid alternative. However, their faster biodegradability to smaller particles (MPs and NPs) poses a potential health risk for animals and humans since they can accumulate in tissues and organs altering homeostasis and physiological functions. Furthermore, bioplastic MPs/NPs are well-studied and investigated as carriers for drug delivery and can easily pass through physiological barriers because they have been developed for these purposes, such as drug loading to assure its proper delivery and improve its bioavailability at the target site.
Despite these bioplastic MPs/NPs exhibit a capacity of complete biodegradation in the body and represent a useful tool for personalized and precision medicine, there are still some issues that should be addressed because side and toxic effects have not been sufficiently investigated, and the majority of data refer to the drug and not to the carriers. Knowing the exact mechanism of distribution, metabolism, and excretion is fundamental to understanding the potential toxic effect of bioplastic MPs/NPs. Until now, there exists a knowledge gap regarding this aspect, and the studies concerning their use for therapeutic purposes are far greater than those aimed at determining their toxicological profile. However, the field of nanotoxicology aimed to understand the interaction between biomaterials and living cells is growing, but studies are still few and methods and results are frequently controversial.
Taken together, all of this evidence emphasizes the importance of the risk assessment for bioplastic MPs/NPs, but it is also necessary to standardize the evaluation method to obtain a reliable validation of their toxicity. Indeed, until now, studies have been conducted using different in vivo and in vitro models, and there are no results about the effects of long-term exposure to these particles. Additionally, because the fabrication of bioplastic MPs/NPs as a delivery system differs based on the method of preparation, there is no general data available about their bio-interaction and biodistribution.

Author Contributions

Conceptualization, A.S., G.D.P., A.V., F.C. and R.M.; funding acquisition, A.S., R.M., F.C., A.T. and G.D.P.; supervision, A.S., A.V., F.C., G.D.P. and R.M.; visualization, E.P.L., M.A.S. and M.M.; writing—original draft, M.M., E.P.L., M.C.C., P.S., M.A.S., R.D., A.T. and N.D.R.; writing—review and editing, E.P.L., M.M., F.C., A.V., R.M., A.T. and A.S. All authors have read and agreed to the published version of the manuscript.

Funding

PRIN PNRR 2022, project code P2022AA47Y to A.S., R.M., F.C. and A.T. and University of Salerno, FARB 2022 to A.S. and G.D.P.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Crutzen, P.J. The “Anthropocene”. In Earth System Science in the Anthropocene; Ehlers, E., Krafft, T., Eds.; Springer-Verlag: Berlin/Heidelberg, Germany, 2006; pp. 13–18. ISBN 978-3-540-26588-7. [Google Scholar]
  2. Zalasiewicz, J.; Waters, C.N.; Ivar Do Sul, J.A.; Corcoran, P.L.; Barnosky, A.D.; Cearreta, A.; Edgeworth, M.; Gałuszka, A.; Jeandel, C.; Leinfelder, R.; et al. The Geological Cycle of Plastics and Their Use as a Stratigraphic Indicator of the Anthropocene. Anthropocene 2016, 13, 4–17. [Google Scholar] [CrossRef]
  3. Minter, A. Junkyard Planet: Travels in the Billion-Dollar Trash Trade, 1st ed.; Bloomsbury Press: New York, NY, USA, 2013; ISBN 978-1-60819-791-0. [Google Scholar]
  4. Hahladakis, J.N.; Velis, C.A.; Weber, R.; Iacovidou, E.; Purnell, P. An Overview of Chemical Additives Present in Plastics: Migration, Release, Fate and Environmental Impact during Their Use, Disposal and Recycling. J. Hazard. Mater. 2018, 344, 179–199. [Google Scholar] [CrossRef] [PubMed]
  5. Sangkham, S.; Faikhaw, O.; Munkong, N.; Sakunkoo, P.; Arunlertaree, C.; Chavali, M.; Mousazadeh, M.; Tiwari, A. A Review on Microplastics and Nanoplastics in the Environment: Their Occurrence, Exposure Routes, Toxic Studies, and Potential Effects on Human Health. Mar. Pollut. Bull. 2022, 181, 113832. [Google Scholar] [CrossRef] [PubMed]
  6. Riechers, M.; Fanini, L.; Apicella, A.; Galván, C.B.; Blondel, E.; Espiña, B.; Kefer, S.; Keroullé, T.; Klun, K.; Pereira, T.R.; et al. Plastics in Our Ocean as Transdisciplinary Challenge. Mar. Pollut. Bull. 2021, 164, 112051. [Google Scholar] [CrossRef]
  7. Narancic, T.; Cerrone, F.; Beagan, N.; O’Connor, K.E. Recent Advances in Bioplastics: Application and Biodegradation. Polymers 2020, 12, 920. [Google Scholar] [CrossRef] [PubMed]
  8. Prajapati, V.D.; Jani, G.K.; Kapadia, J.R. Current Knowledge on Biodegradable Microspheres in Drug Delivery. Expert Opin. Drug Deliv. 2015, 12, 1283–1299. [Google Scholar] [CrossRef]
  9. Su, Y.; Zhang, B.; Sun, R.; Liu, W.; Zhu, Q.; Zhang, X.; Wang, R.; Chen, C. PLGA-Based Biodegradable Microspheres in Drug Delivery: Recent Advances in Research and Application. Drug Deliv. 2021, 28, 1397–1418. [Google Scholar] [CrossRef] [PubMed]
  10. Annu; Sartaj, A.; Qamar, Z.; Md, S.; Alhakamy, N.A.; Baboota, S.; Ali, J. An Insight to Brain Targeting Utilizing Polymeric Nanoparticles: Effective Treatment Modalities for Neurological Disorders and Brain Tumor. Front. Bioeng. Biotechnol. 2022, 10, 788128. [Google Scholar] [CrossRef]
  11. Filiciotto, L.; Rothenberg, G. Biodegradable Plastics: Standards, Policies, and Impacts. ChemSusChem 2021, 14, 56–72. [Google Scholar] [CrossRef]
  12. Wu, Z.; Shi, W.; Valencak, T.G.; Zhang, Y.; Liu, G.; Ren, D. Biodegradation of Conventional Plastics: Candidate Organisms and Potential Mechanisms. Sci. Total Environ. 2023, 885, 163908. [Google Scholar] [CrossRef]
  13. Zimmermann, L.; Dombrowski, A.; Völker, C.; Wagner, M. Are Bioplastics and Plant-Based Materials Safer than Conventional Plastics? In Vitro Toxicity and Chemical Composition. Environ. Int. 2020, 145, 106066. [Google Scholar] [CrossRef]
  14. Rujnić-Sokele, M.; Pilipović, A. Challenges and Opportunities of Biodegradable Plastics: A Mini Review. Waste Manag. Res. 2017, 35, 132–140. [Google Scholar] [CrossRef]
  15. Chandra, R. (Ed.) Environmental Waste Management; CRC Press: Boca Raton, FL, USA, 2016; ISBN 978-1-4987-2475-3. [Google Scholar]
  16. Ghosh, S.K.; Pal, S.; Ray, S. Study of Microbes Having Potentiality for Biodegradation of Plastics. Environ. Sci. Pollut. Res. 2013, 20, 4339–4355. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, X.; Xia, M.; Su, X.; Yuan, P.; Li, X.; Zhou, C.; Wan, Z.; Zou, W. Photolytic Degradation Elevated the Toxicity of Polylactic Acid Microplastics to Developing Zebrafish by Triggering Mitochondrial Dysfunction and Apoptosis. J. Hazard. Mater. 2021, 413, 125321. [Google Scholar] [CrossRef]
  18. Silva, R.R.A.; Marques, C.S.; Arruda, T.R.; Teixeira, S.C.; De Oliveira, T.V. Biodegradation of Polymers: Stages, Measurement, Standards and Prospects. Macromol 2023, 3, 371–399. [Google Scholar] [CrossRef]
  19. Andrady, A.L. The Plastic in Microplastics: A Review. Mar. Pollut. Bull. 2017, 119, 12–22. [Google Scholar] [CrossRef] [PubMed]
  20. Bher, A.; Mayekar, P.C.; Auras, R.A.; Schvezov, C.E. Biodegradation of Biodegradable Polymers in Mesophilic Aerobic Environments. Int. J. Mol. Sci. 2022, 23, 12165. [Google Scholar] [CrossRef]
  21. Lee, W.-T.; Van Muyden, A.; Bobbink, F.D.; Mensi, M.D.; Carullo, J.R.; Dyson, P.J. Mechanistic Classification and Benchmarking of Polyolefin Depolymerization over Silica-Alumina-Based Catalysts. Nat. Commun. 2022, 13, 4850. [Google Scholar] [CrossRef] [PubMed]
  22. Elgharbawy, A.S.; Ali, R.M. A Comprehensive Review of the Polyolefin Composites and Their Properties. Heliyon 2022, 8, e09932. [Google Scholar] [CrossRef]
  23. Hees, T.; Zhong, F.; Stürzel, M.; Mülhaupt, R. Tailoring Hydrocarbon Polymers and All-Hydrocarbon Composites for Circular Economy. Macromol. Rapid Commun. 2019, 40, 1800608. [Google Scholar] [CrossRef]
  24. Yao, Z.; Seong, H.J.; Jang, Y.-S. Environmental Toxicity and Decomposition of Polyethylene. Ecotoxicol. Environ. Saf. 2022, 242, 113933. [Google Scholar] [CrossRef] [PubMed]
  25. Paxton, N.C.; Allenby, M.C.; Lewis, P.M.; Woodruff, M.A. Biomedical Applications of Polyethylene. Eur. Polym. J. 2019, 118, 412–428. [Google Scholar] [CrossRef]
  26. Li, W.C.; Tse, H.F.; Fok, L. Plastic Waste in the Marine Environment: A Review of Sources, Occurrence and Effects. Sci. Total Environ. 2016, 566–567, 333–349. [Google Scholar] [CrossRef]
  27. Galloway, T.S. Micro- and Nano-Plastics and Human Health. In Marine Anthropogenic Litter; Bergmann, M., Gutow, L., Klages, M., Eds.; Springer International Publishing: Cham, Switzerland, 2015; pp. 343–366. ISBN 978-3-319-16509-7. [Google Scholar]
  28. Li, X.; Meng, L.; Zhang, Y.; Qin, Z.; Meng, L.; Li, C.; Liu, M. Research and Application of Polypropylene Carbonate Composite Materials: A Review. Polymers 2022, 14, 2159. [Google Scholar] [CrossRef]
  29. Lewandowski, K.; Skórczewska, K. A Brief Review of Poly(Vinyl Chloride) (PVC) Recycling. Polymers 2022, 14, 3035. [Google Scholar] [CrossRef] [PubMed]
  30. Wypych, G. Chemical Structure of PVC. In PVC Degradation and Stabilization; Elsevier: Amsterdam, The Netherlands, 2015; pp. 1–23. ISBN 978-1-895198-85-0. [Google Scholar]
  31. Abdel-Monem, R.A.; Rabie, S.T.; El-Liethy, M.A.; Hemdan, B.A.; El-Nazer, H.A.; Gaballah, S.T. Chitosan- PVC Conjugates/Metal Nanoparticles for Biomedical Applications. Polym. Adv. Tech. 2022, 33, 514–523. [Google Scholar] [CrossRef]
  32. Wünsch, J.R. Polystyrene: Synthesis, Production and Applications; Rapra Review Reports; Rapra Technology Ltd.: Shawbury, UK, 2000; ISBN 978-1-85957-191-0. [Google Scholar]
  33. Kik, K.; Bukowska, B.; Sicińska, P. Polystyrene Nanoparticles: Sources, Occurrence in the Environment, Distribution in Tissues, Accumulation and Toxicity to Various Organisms. Environ. Pollut. 2020, 262, 114297. [Google Scholar] [CrossRef]
  34. Nisticò, R. Polyethylene Terephthalate (PET) in the Packaging Industry. Polym. Test. 2020, 90, 106707. [Google Scholar] [CrossRef]
  35. Rahman, M.H.; Bhoi, P.R. An Overview of Non-Biodegradable Bioplastics. J. Clean. Prod. 2021, 294, 126218. [Google Scholar] [CrossRef]
  36. Ferreira, F.V.; Cividanes, L.S.; Gouveia, R.F.; Lona, L.M.F. An Overview on Properties and Applications of Poly(Butylene Adipate-co-Terephthalate)-PBAT Based Composites. Polym. Eng. Sci. 2019, 59, E7–E15. [Google Scholar] [CrossRef]
  37. Welle, F. Twenty Years of PET Bottle to Bottle Recycling—An Overview. Resour. Conserv. Recycl. 2011, 55, 865–875. [Google Scholar] [CrossRef]
  38. Chan, D.S.; Fnais, N.; Ibrahim, I.; Daniel, S.J.; Manoukian, J. Exploring Polycaprolactone in Tracheal Surgery: A Scoping Review of in-Vivo Studies. Int. J. Pediatr. Otorhinolaryngol. 2019, 123, 38–42. [Google Scholar] [CrossRef]
  39. Chen, T.; Cai, T.; Jin, Q.; Ji, J. Design and Fabrication of Functional Polycaprolactone. e-Polymers 2015, 15, 3–13. [Google Scholar] [CrossRef]
  40. Hajiali, F.; Tajbakhsh, S.; Shojaei, A. Fabrication and Properties of Polycaprolactone Composites Containing Calcium Phosphate-Based Ceramics and Bioactive Glasses in Bone Tissue Engineering: A Review. Polym. Rev. 2018, 58, 164–207. [Google Scholar] [CrossRef]
  41. Džunuzović, J.V.; Stefanović, I.S.; Džunuzović, E.S.; Dapčević, A.; Šešlija, S.I.; Balanč, B.D.; Lama, G.C. Polyurethane Networks Based on Polycaprolactone and Hyperbranched Polyester: Structural, Thermal and Mechanical Investigation. Prog. Org. Coat. 2019, 137, 105305. [Google Scholar] [CrossRef]
  42. Sadeghi, A.; Mousavi, S.M.; Saljoughi, E.; Kiani, S. Biodegradable Membrane Based on Polycaprolactone/Polybutylene Succinate: Characterization and Performance Evaluation in Wastewater Treatment. J. Appl. Polym. Sci. 2021, 138, 50332. [Google Scholar] [CrossRef]
  43. Lule, Z.C.; Wondu Shiferaw, E.; Kim, J. Thermomechanical Properties of SiC-Filled Polybutylene Succinate Composite Fabricated via Melt Extrusion. Polymers 2020, 12, 418. [Google Scholar] [CrossRef]
  44. Wu, Y.; Gao, X.; Wu, J.; Zhou, T.; Nguyen, T.T.; Wang, Y. Biodegradable Polylactic Acid and Its Composites: Characteristics, Processing, and Sustainable Applications in Sports. Polymers 2023, 15, 3096. [Google Scholar] [CrossRef]
  45. Van Beilen, J.B.; Poirier, Y. Production of Renewable Polymers from Crop Plants. Plant J. 2008, 54, 684–701. [Google Scholar] [CrossRef] [PubMed]
  46. Lasprilla, A.J.R.; Martinez, G.A.R.; Lunelli, B.H.; Jardini, A.L.; Filho, R.M. Poly-Lactic Acid Synthesis for Application in Biomedical Devices—A Review. Biotechnol. Adv. 2012, 30, 321–328. [Google Scholar] [CrossRef]
  47. Garlotta, D. A Literature Review of Poly(Lactic Acid). J. Polym. Environ. 2001, 9, 63–84. [Google Scholar] [CrossRef]
  48. Tait, M.; Pegoretti, A.; Dorigato, A.; Kalaitzidou, K. The Effect of Filler Type and Content and the Manufacturing Process on the Performance of Multifunctional Carbon/Poly-Lactide Composites. Carbon 2011, 49, 4280–4290. [Google Scholar] [CrossRef]
  49. Fambri, L.; Dorigato, A.; Pegoretti, A. Role of Surface-Treated Silica Nanoparticles on the Thermo-Mechanical Behavior of Poly(Lactide). Appl. Sci. 2020, 10, 6731. [Google Scholar] [CrossRef]
  50. Cheng, Y.; Deng, S.; Chen, P.; Ruan, R. Polylactic Acid (PLA) Synthesis and Modifications: A Review. Front. Chem. China 2009, 4, 259–264. [Google Scholar] [CrossRef]
  51. Ahvenainen, R. (Ed.) Novel Food Packaging Techniques; Woodhead Publishing in Food Science and Technology; Woodhead Publishing: Cambridge, UK, 2003; ISBN 978-1-85573-675-7. [Google Scholar]
  52. Jem, K.J.; Tan, B. The Development and Challenges of Poly (Lactic Acid) and Poly (Glycolic Acid). Adv. Ind. Eng. Polym. Res. 2020, 3, 60–70. [Google Scholar] [CrossRef]
  53. Bhatia, S.K.; Otari, S.V.; Jeon, J.-M.; Gurav, R.; Choi, Y.-K.; Bhatia, R.K.; Pugazhendhi, A.; Kumar, V.; Rajesh Banu, J.; Yoon, J.-J.; et al. Biowaste-to-Bioplastic (Polyhydroxyalkanoates): Conversion Technologies, Strategies, Challenges, and Perspective. Bioresour. Technol. 2021, 326, 124733. [Google Scholar] [CrossRef]
  54. Koller, M. Biodegradable and Biocompatible Polyhydroxy-Alkanoates (PHA): Auspicious Microbial Macromolecules for Pharmaceutical and Therapeutic Applications. Molecules 2018, 23, 362. [Google Scholar] [CrossRef] [PubMed]
  55. Kalia, V.C.; Singh Patel, S.K.; Shanmugam, R.; Lee, J.-K. Polyhydroxyalkanoates: Trends and Advances toward Biotechnological Applications. Bioresour. Technol. 2021, 326, 124737. [Google Scholar] [CrossRef]
  56. Silva, J.B.; Pereira, J.R.; Marreiros, B.C.; Reis, M.A.M.; Freitas, F. Microbial Production of Medium-Chain Length Polyhydroxyalkanoates. Process Biochem. 2021, 102, 393–407. [Google Scholar] [CrossRef]
  57. Döhler, N.; Wellenreuther, C.; Wolf, A. Market Dynamics of Biodegradable Bio-Based Plastics: Projections and Linkages to European Policies. EFB Bioecon. J. 2022, 2, 100028. [Google Scholar] [CrossRef]
  58. Fredi, G.; Dorigato, A. Recycling of Bioplastic Waste: A Review. Adv. Ind. Eng. Polym. Res. 2021, 4, 159–177. [Google Scholar] [CrossRef]
  59. Lamberti, F.M.; Román-Ramírez, L.A.; Wood, J. Recycling of Bioplastics: Routes and Benefits. J. Polym. Environ. 2020, 28, 2551–2571. [Google Scholar] [CrossRef]
  60. Cao, G.; Cai, Z. Getting Health Hazards of Inhaled Nano/Microplastics into Focus: Expectations and Challenges. Environ. Sci. Technol. 2023, 57, 3461–3463. [Google Scholar] [CrossRef] [PubMed]
  61. Schneider, M.; Stracke, F.; Hansen, S.; Schaefer, U.F. Nanoparticles and Their Interactions with the Dermal Barrier. Derm. Endocrinol. 2009, 1, 197–206. [Google Scholar] [CrossRef] [PubMed]
  62. Campbell, C.S.J.; Contreras-Rojas, L.R.; Delgado-Charro, M.B.; Guy, R.H. Objective Assessment of Nanoparticle Disposition in Mammalian Skin after Topical Exposure. J. Control. Release 2012, 162, 201–207. [Google Scholar] [CrossRef]
  63. Vogt, A.; Combadiere, B.; Hadam, S.; Stieler, K.M.; Lademann, J.; Schaefer, H.; Autran, B.; Sterry, W.; Blume-Peytavi, U. 40nm, but Not 750 or 1,500nm, Nanoparticles Enter Epidermal CD1a+ Cells after Transcutaneous Application on Human Skin. J. Investig. Dermatol. 2006, 126, 1316–1322. [Google Scholar] [CrossRef]
  64. Zou, Y.; Celli, A.; Zhu, H.; Elmahdy, A.; Cao, Y.; Hui, X.; Maibach, H. Confocal Laser Scanning Microscopy to Estimate Nanoparticles&rsquo; Human Skin Penetration in Vitro. Int. J. Nanomed. 2017, 12, 8035–8041. [Google Scholar] [CrossRef]
  65. Jatana, S.; Callahan, L.; Pentland, A.; DeLouise, L. Impact of Cosmetic Lotions on Nanoparticle Penetration through Ex Vivo C57BL/6 Hairless Mouse and Human Skin: A Comparison Study. Cosmetics 2016, 3, 6. [Google Scholar] [CrossRef]
  66. Alvarez-Román, R.; Naik, A.; Kalia, Y.N.; Guy, R.H.; Fessi, H. Enhancement of Topical Delivery from Biodegradable Nanoparticles. Pharm. Res. 2004, 21, 1818–1825. [Google Scholar] [CrossRef]
  67. Chen, G.; Feng, Q.; Wang, J. Mini-Review of Microplastics in the Atmosphere and Their Risks to Humans. Sci. Total Environ. 2020, 703, 135504. [Google Scholar] [CrossRef]
  68. Dris, R.; Gasperi, J.; Mirande, C.; Mandin, C.; Guerrouache, M.; Langlois, V.; Tassin, B. A First Overview of Textile Fibers, Including Microplastics, in Indoor and Outdoor Environments. Environ. Pollut. 2017, 221, 453–458. [Google Scholar] [CrossRef]
  69. Stapleton, P. Toxicological Considerations of Nano-Sized Plastics. AIMS Environ. Sci. 2019, 6, 367–378. [Google Scholar] [CrossRef]
  70. Prata, J.C. Airborne Microplastics: Consequences to Human Health? Environ. Pollut. 2018, 234, 115–126. [Google Scholar] [CrossRef]
  71. Pauly, J.L.; Stegmeier, S.J.; Allaart, H.A.; Cheney, R.T.; Zhang, P.J.; Mayer, A.G.; Streck, R.J. Inhaled Cellulosic and Plastic Fibers Found in Human Lung Tissue. Cancer Epidemiol. Biomark. Prev. 1998, 7, 419–428. [Google Scholar]
  72. Dong, C.-D.; Chen, C.-W.; Chen, Y.-C.; Chen, H.-H.; Lee, J.-S.; Lin, C.-H. Polystyrene Microplastic Particles: In Vitro Pulmonary Toxicity Assessment. J. Hazard. Mater. 2020, 385, 121575. [Google Scholar] [CrossRef] [PubMed]
  73. Brown, D.M.; Wilson, M.R.; MacNee, W.; Stone, V.; Donaldson, K. Size-Dependent Proinflammatory Effects of Ultrafine Polystyrene Particles: A Role for Surface Area and Oxidative Stress in the Enhanced Activity of Ultrafines. Toxicol. Appl. Pharmacol. 2001, 175, 191–199. [Google Scholar] [CrossRef]
  74. Bengalli, R.; Zerboni, A.; Bonfanti, P.; Saibene, M.; Mehn, D.; Cella, C.; Ponti, J.; La Spina, R.; Mantecca, P. Characterization of Microparticles Derived from Waste Plastics and Their Bio-interaction with Human Lung A549 Cells. J. Appl. Toxicol. 2022, 42, 2030–2044. [Google Scholar] [CrossRef] [PubMed]
  75. Wu, H.; Liu, Q.; Yang, N.; Xu, S. Polystyrene-Microplastics and DEHP Co-Exposure Induced DNA Damage, Cell Cycle Arrest and Necroptosis of Ovarian Granulosa Cells in Mice by Promoting ROS Production. Sci. Total Environ. 2023, 871, 161962. [Google Scholar] [CrossRef] [PubMed]
  76. Li, Y.; Shi, T.; Li, X.; Sun, H.; Xia, X.; Ji, X.; Zhang, J.; Liu, M.; Lin, Y.; Zhang, R.; et al. Inhaled Tire-Wear Microplastic Particles Induced Pulmonary Fibrotic Injury via Epithelial Cytoskeleton Rearrangement. Environ. Int. 2022, 164, 107257. [Google Scholar] [CrossRef] [PubMed]
  77. Lehner, R.; Weder, C.; Petri-Fink, A.; Rothen-Rutishauser, B. Emergence of Nanoplastic in the Environment and Possible Impact on Human Health. Environ. Sci. Technol. 2019, 53, 1748–1765. [Google Scholar] [CrossRef] [PubMed]
  78. Eyles, J.E.; Bramwell, V.W.; Williamson, E.D.; Alpar, H.O. Microsphere Translocation and Immunopotentiation in Systemic Tissues Following Intranasal Administration. Vaccine 2001, 19, 4732–4742. [Google Scholar] [CrossRef]
  79. Islam, S.U.; Shehzad, A.; Ahmed, M.B.; Lee, Y.S. Intranasal Delivery of Nanoformulations: A Potential Way of Treatment for Neurological Disorders. Molecules 2020, 25, 1929. [Google Scholar] [CrossRef] [PubMed]
  80. Oberdörster, G.; Sharp, Z.; Atudorei, V.; Elder, A.; Gelein, R.; Kreyling, W.; Cox, C. Translocation of Inhaled Ultrafine Particles to the Brain. Inhal. Toxicol. 2004, 16, 437–445. [Google Scholar] [CrossRef]
  81. Elder, A.; Gelein, R.; Silva, V.; Feikert, T.; Opanashuk, L.; Carter, J.; Potter, R.; Maynard, A.; Ito, Y.; Finkelstein, J.; et al. Translocation of Inhaled Ultrafine Manganese Oxide Particles to the CentralNervous System. Environ. Health Perspect. 2006, 114, 1172–1178. [Google Scholar] [CrossRef] [PubMed]
  82. Qi, Y.; Wei, S.; Xin, T.; Huang, C.; Pu, Y.; Ma, J.; Zhang, C.; Liu, Y.; Lynch, I.; Liu, S. Passage of Exogeneous Fine Particles from the Lung into the Brain in Humans and Animals. Proc. Natl. Acad. Sci. USA 2022, 119, e2117083119. [Google Scholar] [CrossRef] [PubMed]
  83. Alvarez-Román, R.; Naik, A.; Kalia, Y.N.; Guy, R.H.; Fessi, H. Skin Penetration and Distribution of Polymeric Nanoparticles. J. Control. Release 2004, 99, 53–62. [Google Scholar] [CrossRef]
  84. Karami, A.; Golieskardi, A.; Ho, Y.B.; Larat, V.; Salamatinia, B. Microplastics in Eviscerated Flesh and Excised Organs of Dried Fish. Sci. Rep. 2017, 7, 5473. [Google Scholar] [CrossRef]
  85. Li, B.; Ding, Y.; Cheng, X.; Sheng, D.; Xu, Z.; Rong, Q.; Wu, Y.; Zhao, H.; Ji, X.; Zhang, Y. Polyethylene Microplastics Affect the Distribution of Gut Microbiota and Inflammation Development in Mice. Chemosphere 2020, 244, 125492. [Google Scholar] [CrossRef] [PubMed]
  86. Xu, D.; Ma, Y.; Han, X.; Chen, Y. Systematic Toxicity Evaluation of Polystyrene Nanoplastics on Mice and Molecular Mechanism Investigation about Their Internalization into Caco-2 Cells. J. Hazard. Mater. 2021, 417, 126092. [Google Scholar] [CrossRef]
  87. Han, J.; Yan, J.; Li, K.; Lin, B.; Lai, W.; Bian, L.; Jia, R.; Liu, X.; Xi, Z. Distribution of Micro-Nano PS, DEHP, and/or MEHP in Mice and Nerve Cell Models In Vitro after Exposure to Micro-Nano PS and DEHP. Toxics 2023, 11, 441. [Google Scholar] [CrossRef] [PubMed]
  88. Güven, O.; Gökdağ, K.; Jovanović, B.; Kıdeyş, A.E. Microplastic Litter Composition of the Turkish Territorial Waters of the Mediterranean Sea, and Its Occurrence in the Gastrointestinal Tract of Fish. Environ. Pollut. 2017, 223, 286–294. [Google Scholar] [CrossRef]
  89. Banerjee, A.; Billey, L.O.; Shelver, W.L. Uptake and Toxicity of Polystyrene Micro/Nanoplastics in Gastric Cells: Effects of Particle Size and Surface Functionalization. PLoS ONE 2021, 16, e0260803. [Google Scholar] [CrossRef] [PubMed]
  90. Domenech, J.; De Britto, M.; Velázquez, A.; Pastor, S.; Hernández, A.; Marcos, R.; Cortés, C. Long-Term Effects of Polystyrene Nanoplastics in Human Intestinal Caco-2 Cells. Biomolecules 2021, 11, 1442. [Google Scholar] [CrossRef]
  91. Domenech, J.; Hernández, A.; Rubio, L.; Marcos, R.; Cortés, C. Interactions of Polystyrene Nanoplastics with in Vitro Models of the Human Intestinal Barrier. Arch. Toxicol. 2020, 94, 2997–3012. [Google Scholar] [CrossRef]
  92. Shan, S.; Zhang, Y.; Zhao, H.; Zeng, T.; Zhao, X. Polystyrene Nanoplastics Penetrate across the Blood-Brain Barrier and Induce Activation of Microglia in the Brain of Mice. Chemosphere 2022, 298, 134261. [Google Scholar] [CrossRef]
  93. Yang, D.; Zhu, J.; Zhou, X.; Pan, D.; Nan, S.; Yin, R.; Lei, Q.; Ma, N.; Zhu, H.; Chen, J.; et al. Polystyrene Micro- and Nano-Particle Coexposure Injures Fetal Thalamus by Inducing ROS-Mediated Cell Apoptosis. Environ. Int. 2022, 166, 107362. [Google Scholar] [CrossRef]
  94. Ciaglia, E.; Montella, F.; Trucillo, P.; Ciardulli, M.C.; Di Pietro, P.; Amodio, G.; Remondelli, P.; Vecchione, C.; Reverchon, E.; Maffulli, N.; et al. A Bioavailability Study on Microbeads and Nanoliposomes Fabricated by Dense Carbon Dioxide Technologies Using Human-Primary Monocytes and Flow Cytometry Assay. Int. J. Pharm. 2019, 570, 118686. [Google Scholar] [CrossRef]
  95. Govoni, M.; Lamparelli, E.P.; Ciardulli, M.C.; Santoro, A.; Oliviero, A.; Palazzo, I.; Reverchon, E.; Vivarelli, L.; Maso, A.; Storni, E.; et al. Demineralized Bone Matrix Paste Formulated with Biomimetic PLGA Microcarriers for the Vancomycin Hydrochloride Controlled Delivery: Release Profile, Citotoxicity and Efficacy against S. Aureus. Int. J. Pharm. 2020, 582, 119322. [Google Scholar] [CrossRef] [PubMed]
  96. Palazzo, I.; Lamparelli, E.P.; Ciardulli, M.C.; Scala, P.; Reverchon, E.; Forsyth, N.; Maffulli, N.; Santoro, A.; Della Porta, G. Supercritical Emulsion Extraction Fabricated PLA/PLGA Micro/Nano Carriers for Growth Factor Delivery: Release Profiles and Cytotoxicity. Int. J. Pharm. 2021, 592, 120108. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, Q.; Chen, G.; Tian, L.; Kong, C.; Gao, D.; Chen, Y.; Junaid, M.; Wang, J. Neuro- and Hepato-Toxicity of Polystyrene Nanoplastics and Polybrominated Diphenyl Ethers on Early Life Stages of Zebrafish. Sci. Total Environ. 2023, 857, 159567. [Google Scholar] [CrossRef]
  98. Kumari, A.; Yadav, S.K.; Yadav, S.C. Biodegradable Polymeric Nanoparticles Based Drug Delivery Systems. Colloids Surf. B Biointerfaces 2010, 75, 1–18. [Google Scholar] [CrossRef] [PubMed]
  99. Zhong, H.; Chan, G.; Hu, Y.; Hu, H.; Ouyang, D. A Comprehensive Map of FDA-Approved Pharmaceutical Products. Pharmaceutics 2018, 10, 263. [Google Scholar] [CrossRef]
  100. Ghitman, J.; Biru, E.I.; Stan, R.; Iovu, H. Review of Hybrid PLGA Nanoparticles: Future of Smart Drug Delivery and Theranostics Medicine. Mater. Des. 2020, 193, 108805. [Google Scholar] [CrossRef]
  101. Alsaab, H.O.; Alharbi, F.D.; Alhibs, A.S.; Alanazi, N.B.; Alshehri, B.Y.; Saleh, M.A.; Alshehri, F.S.; Algarni, M.A.; Almugaiteeb, T.; Uddin, M.N.; et al. PLGA-Based Nanomedicine: History of Advancement and Development in Clinical Applications of Multiple Diseases. Pharmaceutics 2022, 14, 2728. [Google Scholar] [CrossRef] [PubMed]
  102. Akhtar, A.; Andleeb, A.; Waris, T.S.; Bazzar, M.; Moradi, A.-R.; Awan, N.R.; Yar, M. Neurodegenerative Diseases and Effective Drug Delivery: A Review of Challenges and Novel Therapeutics. J. Control. Release 2021, 330, 1152–1167. [Google Scholar] [CrossRef]
  103. Vert, M.; Doi, Y.; Hellwich, K.-H.; Hess, M.; Hodge, P.; Kubisa, P.; Rinaudo, M.; Schué, F. Terminology for Biorelated Polymers and Applications (IUPAC Recommendations 2012). Pure Appl. Chem. 2012, 84, 377–410. [Google Scholar] [CrossRef]
  104. Faiyaz, M. Nanomaterials in Alzheimer’s Disease Treatment: A Comprehensive Review. Front. Biosci. 2021, 26, 851. [Google Scholar] [CrossRef]
  105. Anselmo, A.C.; Mitragotri, S. An Overview of Clinical and Commercial Impact of Drug Delivery Systems. J. Control. Release 2014, 190, 15–28. [Google Scholar] [CrossRef]
  106. Molavi, F.; Barzegar-Jalali, M.; Hamishehkar, H. Polyester Based Polymeric Nano and Microparticles for Pharmaceutical Purposes: A Review on Formulation Approaches. J. Control. Release 2020, 320, 265–282. [Google Scholar] [CrossRef]
  107. Tawde, V.; Chaurasia, S.; Gupta, S.; Rastogi, R.; Dantuluri, A.K.; Liu, W.; Mcmahon, S.; Seo, D.-W. Smart Bioreso Rbable Polymers for Pharmaceuticals and Medical Devices. Yakhak Hoeji 2022, 66, 1–6. [Google Scholar] [CrossRef]
  108. Mahalingam, M.; Krishnamoorthy, K. Selection of a Suitable Method for the Preparation of Polymeric Nanoparticles: Multi-Criteria Decision Making Approach. Adv. Pharm. Bull. 2015, 5, 57. [Google Scholar] [CrossRef]
  109. Zielińska, A.; Carreiró, F.; Oliveira, A.M.; Neves, A.; Pires, B.; Venkatesh, D.N.; Durazzo, A.; Lucarini, M.; Eder, P.; Silva, A.M.; et al. Polymeric Nanoparticles: Production, Characterization, Toxicology and Ecotoxicology. Molecules 2020, 25, 3731. [Google Scholar] [CrossRef]
  110. Iván Martínez-Muñoz, O.; Fernando Ospina-Giraldo, L.; Elizabeth Mora-Huertas, C. Nanoprecipitation: Applications for Entrapping Active Molecules of Interest in Pharmaceutics. In Nano- and Microencapsulation—Techniques and Applications; Abu-Thabit, N., Ed.; IntechOpen: London, UK, 2021; ISBN 978-1-83968-348-0. [Google Scholar]
  111. Bilati, U.; Allémann, E.; Doelker, E. Development of a Nanoprecipitation Method Intended for the Entrapment of Hydrophilic Drugs into Nanoparticles. Eur. J. Pharm. Sci. 2005, 24, 67–75. [Google Scholar] [CrossRef] [PubMed]
  112. Barichello, J.M.; Morishita, M.; Takayama, K.; Nagai, T. Encapsulation of Hydrophilic and Lipophilic Drugs in PLGA Nanoparticles by the Nanoprecipitation Method. Drug Dev. Ind. Pharm. 1999, 25, 471–476. [Google Scholar] [CrossRef] [PubMed]
  113. Leung, M.H.M.; Shen, A.Q. Microfluidic Assisted Nanoprecipitation of PLGA Nanoparticles for Curcumin Delivery to Leukemia Jurkat Cells. Langmuir 2018, 34, 3961–3970. [Google Scholar] [CrossRef]
  114. Lamparelli, E.P.; Ciardulli, M.C.; Scala, P.; Scognamiglio, M.; Charlier, B.; Di Pietro, P.; Izzo, V.; Vecchione, C.; Maffulli, N.; Della Porta, G. Lipid Nano-Vesicles for Thyroid Hormone Encapsulation: A Comparison between Different Fabrication Technologies, Drug Loading, and an in Vitro Delivery to Human Tendon Stem/Progenitor Cells in 2D and 3D Culture. Int. J. Pharm. 2022, 624, 122007. [Google Scholar] [CrossRef]
  115. Zhang, H.; Yang, J.; Sun, R.; Han, S.; Yang, Z.; Teng, L. Microfluidics for Nano-Drug Delivery Systems: From Fundamentals to Industrialization. Acta Pharm. Sin. B 2023, 13, 3277–3299. [Google Scholar] [CrossRef] [PubMed]
  116. Freitas, S.; Merkle, H.P.; Gander, B. Microencapsulation by Solvent Extraction/Evaporation: Reviewing the State of the Art of Microsphere Preparation Process Technology. J. Control. Release 2005, 102, 313–332. [Google Scholar] [CrossRef] [PubMed]
  117. Katou, H.; Wandrey, A.J.; Gander, B. Kinetics of Solvent Extraction/Evaporation Process for PLGA Microparticle Fabrication. Int. J. Pharm. 2008, 364, 45–53. [Google Scholar] [CrossRef]
  118. Nag, D.; Nath, B. Nath Review on Solvent Evaporation Technique: A Promising Method for Microencapsulation. World J. Pharm. Res. 2018, 7, 356–372. [Google Scholar]
  119. Meng, F.T.; Ma, G.H.; Liu, Y.D.; Qiu, W.; Su, Z.G. Microencapsulation of Bovine Hemoglobin with High Bio-Activity and High Entrapment Efficiency Using a W/O/W Double Emulsion Technique. Colloids Surf. B Biointerfaces 2004, 33, 177–183. [Google Scholar] [CrossRef]
  120. Miyazaki, Y.; Onuki, Y.; Yakou, S.; Takayama, K. Effect of Temperature-Increase Rate on Drug Release Characteristics of Dextran Microspheres Prepared by Emulsion Solvent Evaporation Process. Int. J. Pharm. 2006, 324, 144–151. [Google Scholar] [CrossRef] [PubMed]
  121. Ozkan, G.; Franco, P.; De Marco, I.; Xiao, J.; Capanoglu, E. A Review of Microencapsulation Methods for Food Antioxidants: Principles, Advantages, Drawbacks and Applications. Food Chem. 2019, 272, 494–506. [Google Scholar] [CrossRef] [PubMed]
  122. Della Porta, G.; Adami, R.; Gaudio, P.D.; Prota, L.; Aquino, R.; Reverchon, E. Albumin/Gentamicin Microspheres Produced by Supercritical Assisted Atomization: Optimization of Size, Drug Loading and Release. J. Pharm. Sci. 2010, 99, 4720–4729. [Google Scholar] [CrossRef] [PubMed]
  123. Della Porta, G.; De Vittori, C.; Reverchon, E. Supercritical Assisted Atomization: A Novel Technology for Microparticles Preparation of an Asthma-Controlling Drug. AAPS PharmSciTech 2005, 6, E421–E428. [Google Scholar] [CrossRef]
  124. Campardelli, R.; Della Porta, G.; Reverchon, E. Solvent Elimination from Polymer Nanoparticle Suspensions by Continuous Supercritical Extraction. J. Supercrit. Fluids 2012, 70, 100–105. [Google Scholar] [CrossRef]
  125. Campardelli, R.; Adami, R.; Della Porta, G.; Reverchon, E. Nanoparticle Precipitation by Supercritical Assisted Injection in a Liquid Antisolvent. Chem. Eng. J. 2012, 192, 246–251. [Google Scholar] [CrossRef]
  126. Trucillo, E.; Bisceglia, B.; Valdrè, G.; Giordano, E.; Reverchon, E.; Maffulli, N.; Della Porta, G. Growth Factor Sustained Delivery from Poly-lactic-co-glycolic Acid Microcarriers and Its Mass Transfer Modeling by Finite Element in a Dynamic and Static Three-dimensional Environment Bioengineered with Stem Cells. Biotech. Bioeng. 2019, 116, 1777–1794. [Google Scholar] [CrossRef] [PubMed]
  127. Della Porta, G.; Falco, N.; Giordano, E.; Reverchon, E. PLGA Microspheres by Supercritical Emulsion Extraction: A Study on Insulin Release in Myoblast Culture. J. Biomater. Sci. Polym. Ed. 2013, 24, 1831–1847. [Google Scholar] [CrossRef]
  128. Della Porta, G.; Campardelli, R.; Falco, N.; Reverchon, E. PLGA Microdevices for Retinoids Sustained Release Produced by Supercritical Emulsion Extraction: Continuous versus Batch Operation Layouts. J. Pharm. Sci. 2011, 100, 4357–4367. [Google Scholar] [CrossRef]
  129. Falco, N.; Reverchon, E.; Della Porta, G. Injectable PLGA/Hydrocortisone Formulation Produced by Continuous Supercritical Emulsion Extraction. Int. J. Pharm. 2013, 441, 589–597. [Google Scholar] [CrossRef] [PubMed]
  130. Cricchio, V.; Best, M.; Reverchon, E.; Maffulli, N.; Phillips, G.; Santin, M.; Della Porta, G. Novel Superparamagnetic Microdevices Based on Magnetized PLGA/PLA Microparticles Obtained by Supercritical Fluid Emulsion and Coating by Carboxybetaine-Functionalized Chitosan Allowing the Tuneable Release of Therapeutics. J. Pharm. Sci. 2017, 106, 2097–2105. [Google Scholar] [CrossRef] [PubMed]
  131. Palazzo, I.; Raimondo, M.; Della Porta, G.; Guadagno, L.; Reverchon, E. Encapsulation of Health-Monitoring Agent in Poly-Methyl-Methacrylate Microcapsules Using Supercritical Emulsion Extraction. J. Ind. Eng. Chem. 2020, 90, 287–299. [Google Scholar] [CrossRef]
  132. Morais, A.Í.S.; Vieira, E.G.; Afewerki, S.; Sousa, R.B.; Honorio, L.M.C.; Cambrussi, A.N.C.O.; Santos, J.A.; Bezerra, R.D.S.; Furtini, J.A.O.; Silva-Filho, E.C.; et al. Fabrication of Polymeric Microparticles by Electrospray: The Impact of Experimental Parameters. J. Funct. Biomater. 2020, 11, 4. [Google Scholar] [CrossRef] [PubMed]
  133. Strojewski, D.; Krupa, A. Spray Drying and Nano Spray Drying as Manufacturing Methods of Drug-Loaded Polymeric Particles. Polim. Med. 2022, 52, 101–111. [Google Scholar] [CrossRef]
  134. Adepu, S.; Ramakrishna, S. Controlled Drug Delivery Systems: Current Status and Future Directions. Molecules 2021, 26, 5905. [Google Scholar] [CrossRef]
  135. Hanson, L.R.; Frey, W.H. Intranasal Delivery Bypasses the Blood-Brain Barrier to Target Therapeutic Agents to the Central Nervous System and Treat Neurodegenerative Disease. BMC Neurosci. 2008, 9, S5. [Google Scholar] [CrossRef]
  136. Patel, T.; Zhou, J.; Piepmeier, J.M.; Saltzman, W.M. Polymeric Nanoparticles for Drug Delivery to the Central Nervous System. Adv. Drug Deliv. Rev. 2012, 64, 701–705. [Google Scholar] [CrossRef]
  137. Fowler, M.J.; Cotter, J.D.; Knight, B.E.; Sevick-Muraca, E.M.; Sandberg, D.I.; Sirianni, R.W. Intrathecal Drug Delivery in the Era of Nanomedicine. Adv. Drug Deliv. Rev. 2020, 165–166, 77–95. [Google Scholar] [CrossRef]
  138. Bukari, B.; Samarasinghe, R.M.; Noibanchong, J.; Shigdar, S.L. Non-Invasive Delivery of Therapeutics into the Brain: The Potential of Aptamers for Targeted Delivery. Biomedicines 2020, 8, 120. [Google Scholar] [CrossRef]
  139. Burgess, A.; Shah, K.; Hough, O.; Hynynen, K. Focused Ultrasound-Mediated Drug Delivery through the Blood–Brain Barrier. Expert Rev. Neurother. 2015, 15, 477–491. [Google Scholar] [CrossRef]
  140. Pinheiro, R.G.R.; Coutinho, A.J.; Pinheiro, M.; Neves, A.R. Nanoparticles for Targeted Brain Drug Delivery: What Do We Know? Int. J. Mol. Sci. 2021, 22, 11654. [Google Scholar] [CrossRef]
  141. Hersh, A.M.; Alomari, S.; Tyler, B.M. Crossing the Blood-Brain Barrier: Advances in Nanoparticle Technology for Drug Delivery in Neuro-Oncology. Int. J. Mol. Sci. 2022, 23, 4153. [Google Scholar] [CrossRef]
  142. Kou, L.; Bhutia, Y.D.; Yao, Q.; He, Z.; Sun, J.; Ganapathy, V. Transporter-Guided Delivery of Nanoparticles to Improve Drug Permeation across Cellular Barriers and Drug Exposure to Selective Cell Types. Front. Pharmacol. 2018, 9, 27. [Google Scholar] [CrossRef]
  143. Zhang, S.; Gan, L.; Cao, F.; Wang, H.; Gong, P.; Ma, C.; Ren, L.; Lin, Y.; Lin, X. The Barrier and Interface Mechanisms of the Brain Barrier, and Brain Drug Delivery. Brain Res. Bull. 2022, 190, 69–83. [Google Scholar] [CrossRef]
  144. Botti, G.; Dalpiaz, A.; Pavan, B. Targeting Systems to the Brain Obtained by Merging Prodrugs, Nanoparticles, and Nasal Administration. Pharmaceutics 2021, 13, 1144. [Google Scholar] [CrossRef] [PubMed]
  145. Yusuf, M.; Khan, M.; Alrobaian, M.M.; Alghamdi, S.A.; Warsi, M.H.; Sultana, S.; Khan, R.A. Brain Targeted Polysorbate-80 Coated PLGA Thymoquinone Nanoparticles for the Treatment of Alzheimer’s Disease, with Biomechanistic Insights. J. Drug Deliv. Sci. Technol. 2021, 61, 102214. [Google Scholar] [CrossRef]
  146. Elibol, B.; Beker, M.; Terzioglu-Usak, S.; Dalli, T.; Kilic, U. Thymoquinone Administration Ameliorates Alzheimer’s Disease-like Phenotype by Promoting Cell Survival in the Hippocampus of Amyloid Beta1–42 Infused Rat Model. Phytomedicine 2020, 79, 153324. [Google Scholar] [CrossRef]
  147. Xu, R.; Wang, J.; Xu, J.; Song, X.; Huang, H.; Feng, Y.; Fu, C. Rhynchophylline Loaded-mPEG-PLGA Nanoparticles Coated with Tween-80 for Preliminary Study in Alzheimer’s Disease. Int. J. Nanomed. 2020, 15, 1149–1160. [Google Scholar] [CrossRef] [PubMed]
  148. Shao, H.; Mi, Z.; Ji, W.; Zhang, C.; Zhang, T.; Ren, S.; Zhu, Z. Rhynchophylline Protects Against the Amyloid β-Induced Increase of Spontaneous Discharges in the Hippocampal CA1 Region of Rats. Neurochem. Res. 2015, 40, 2365–2373. [Google Scholar] [CrossRef]
  149. Carradori, D.; Balducci, C.; Re, F.; Brambilla, D.; Le Droumaguet, B.; Flores, O.; Gaudin, A.; Mura, S.; Forloni, G.; Ordoñez-Gutierrez, L.; et al. Antibody-Functionalized Polymer Nanoparticle Leading to Memory Recovery in Alzheimer’s Disease-like Transgenic Mouse Model. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 609–618. [Google Scholar] [CrossRef] [PubMed]
  150. Duty, S.; Jenner, P. Animal Models of Parkinson’s Disease: A Source of Novel Treatments and Clues to the Cause of the Disease: Animal Models of Parkinson’s Disease. Br. J. Pharmacol. 2011, 164, 1357–1391. [Google Scholar] [CrossRef] [PubMed]
  151. De Virgilio, A.; Greco, A.; Fabbrini, G.; Inghilleri, M.; Rizzo, M.I.; Gallo, A.; Conte, M.; Rosato, C.; Ciniglio Appiani, M.; De Vincentiis, M. Parkinson’s Disease: Autoimmunity and Neuroinflammation. Autoimmun. Rev. 2016, 15, 1005–1011. [Google Scholar] [CrossRef]
  152. Bordoni, M.; Scarian, E.; Rey, F.; Gagliardi, S.; Carelli, S.; Pansarasa, O.; Cereda, C. Biomaterials in Neurodegenerative Disorders: A Promising Therapeutic Approach. IJMS 2020, 21, 3243. [Google Scholar] [CrossRef] [PubMed]
  153. Ellis, J.M.; Fell, M.J. Current Approaches to the Treatment of Parkinson’s Disease. Bioorg. Med. Chem. Lett. 2017, 27, 4247–4255. [Google Scholar] [CrossRef]
  154. Monge-Fuentes, V.; Biolchi Mayer, A.; Lima, M.R.; Geraldes, L.R.; Zanotto, L.N.; Moreira, K.G.; Martins, O.P.; Piva, H.L.; Felipe, M.S.S.; Amaral, A.C.; et al. Dopamine-Loaded Nanoparticle Systems Circumvent the Blood–Brain Barrier Restoring Motor Function in Mouse Model for Parkinson’s Disease. Sci. Rep. 2021, 11, 15185. [Google Scholar] [CrossRef]
  155. Arisoy, S.; Sayiner, O.; Comoglu, T.; Onal, D.; Atalay, O.; Pehlivanoglu, B. In Vitro and in Vivo Evaluation of Levodopa-Loaded Nanoparticles for Nose to Brain Delivery. Pharm. Dev. Technol. 2020, 25, 735–747. [Google Scholar] [CrossRef] [PubMed]
  156. Higazy, I.M. Brain Targeting Stealth Lipomers of Combined Antiepileptic-Anti-Inflammatory Drugs as Alternative Therapy for Conventional Anti-Parkinson’s. Saudi Pharm. J. 2020, 28, 33–57. [Google Scholar] [CrossRef]
  157. Jarosińska, O.D.; Rüdiger, S.G.D. Molecular Strategies to Target Protein Aggregation in Huntington’s Disease. Front. Mol. Biosci. 2021, 8, 769184. [Google Scholar] [CrossRef] [PubMed]
  158. Caron, N.S.; Haqqani, A.S.; Sandhu, A.; Aly, A.E.; Findlay Black, H.; Bone, J.N.; McBride, J.L.; Abulrob, A.; Stanimirovic, D.; Leavitt, B.R.; et al. Cerebrospinal Fluid Biomarkers for Assessing Huntington Disease Onset and Severity. Brain Commun. 2022, 4, fcac309. [Google Scholar] [CrossRef]
  159. Joshi, A.S.; Singh, V.; Gahane, A.; Thakur, A.K. Biodegradable Nanoparticles Containing Mechanism Based Peptide Inhibitors Reduce Polyglutamine Aggregation in Cell Models and Alleviate Motor Symptoms in a Drosophila Model of Huntington’s Disease. ACS Chem. Neurosci. 2019, 10, 1603–1614. [Google Scholar] [CrossRef]
  160. Leoni, V.; Caccia, C. The Impairment of Cholesterol Metabolism in Huntington Disease. Biochim. Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2015, 1851, 1095–1105. [Google Scholar] [CrossRef] [PubMed]
  161. Kolarcik, C.L.; Bowser, R. Retinoid Signaling Alterations in Amyotrophic Lateral Sclerosis. Am. J. Neurodegener. Dis. 2012, 1, 130–145. [Google Scholar] [CrossRef]
  162. Medina, D.X.; Chung, E.P.; Teague, C.D.; Bowser, R.; Sirianni, R.W. Intravenously Administered, Retinoid Activating Nanoparticles Increase Lifespan and Reduce Neurodegeneration in the SOD1G93A Mouse Model of ALS. Front. Bioeng. Biotechnol. 2020, 8, 224. [Google Scholar] [CrossRef]
  163. Baecher-Allan, C.; Kaskow, B.J.; Weiner, H.L. Multiple Sclerosis: Mechanisms and Immunotherapy. Neuron 2018, 97, 742–768. [Google Scholar] [CrossRef]
  164. Gholamzad, M.; Baharlooi, H.; Shafiee Ardestani, M.; Seyedkhan, Z.; Azimi, M. Prophylactic and Therapeutic Effects of MOG-Conjugated PLGA Nanoparticles in C57Bl/6 Mouse Model of Multiple Sclerosis. Adv. Pharm. Bull. 2020, 11, 505–513. [Google Scholar] [CrossRef] [PubMed]
  165. Haque, S.; Norbert, C.C.; Patra, C.R. Nanomedicine: Future Therapy for Brain Cancers. In Nano Drug Delivery Strategies for the Treatment of Cancers; Elsevier: Amsterdam, The Netherlands, 2021; pp. 37–74. ISBN 978-0-12-819793-6. [Google Scholar]
  166. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  167. Maksimenko, O.; Malinovskaya, J.; Shipulo, E.; Osipova, N.; Razzhivina, V.; Arantseva, D.; Yarovaya, O.; Mostovaya, U.; Khalansky, A.; Fedoseeva, V.; et al. Doxorubicin-Loaded PLGA Nanoparticles for the Chemotherapy of Glioblastoma: Towards the Pharmaceutical Development. Int. J. Pharm. 2019, 572, 118733. [Google Scholar] [CrossRef] [PubMed]
  168. Cui, Y.; Xu, Q.; Chow, P.K.-H.; Wang, D.; Wang, C.-H. Transferrin-Conjugated Magnetic Silica PLGA Nanoparticles Loaded with Doxorubicin and Paclitaxel for Brain Glioma Treatment. Biomaterials 2013, 34, 8511–8520. [Google Scholar] [CrossRef]
  169. Agarwal, S.; Mohamed, M.S.; Mizuki, T.; Maekawa, T.; Sakthi Kumar, D. Chlorotoxin Modified Morusin–PLGA Nanoparticles for Targeted Glioblastoma Therapy. J. Mater. Chem. B 2019, 7, 5896–5919. [Google Scholar] [CrossRef] [PubMed]
  170. Cano, A.; Sánchez-López, E.; Ettcheto, M.; López-Machado, A.; Espina, M.; Souto, E.B.; Galindo, R.; Camins, A.; García, M.L.; Turowski, P. Current Advances in the Development of Novel Polymeric Nanoparticles for the Treatment of Neurodegenerative Diseases. Nanomedicine 2020, 15, 1239–1261. [Google Scholar] [CrossRef] [PubMed]
  171. Tang, H.; Feng, X.; Zhang, T.; Dai, Y.; Zhou, Z.; Chen, H.; Liu, L.; Li, X.; Zhuang, T.; Liu, X.; et al. Stability, Pharmacokinetics, Biodistribution and Safety Assessment of Folate-Conjugated Pullulan Acetate Nanoparticles as Cervical Cancer Targeted Drug Carriers. J. Nanosci. Nanotechnol. 2015, 15, 6405–6412. [Google Scholar] [CrossRef]
  172. Missaoui, W.N.; Arnold, R.D.; Cummings, B.S. Toxicological Status of Nanoparticles: What We Know and What We Don’t Know. Chem.-Biol. Interact. 2018, 295, 1–12. [Google Scholar] [CrossRef] [PubMed]
  173. Dhas, N.; Mehta, T. Cationic Biopolymer Functionalized Nanoparticles Encapsulating Lutein to Attenuate Oxidative Stress in Effective Treatment of Alzheimer’s Disease: A Non-Invasive Approach. Int. J. Pharm. 2020, 586, 119553. [Google Scholar] [CrossRef] [PubMed]
  174. Leite, P.E.C.; Pereira, M.R.; Harris, G.; Pamies, D.; Dos Santos, L.M.G.; Granjeiro, J.M.; Hogberg, H.T.; Hartung, T.; Smirnova, L. Suitability of 3D Human Brain Spheroid Models to Distinguish Toxic Effects of Gold and Poly-Lactic Acid Nanoparticles to Assess Biocompatibility for Brain Drug Delivery. Part. Fibre Toxicol. 2019, 16, 22. [Google Scholar] [CrossRef] [PubMed]
  175. Win-Shwe, T.-T.; Fujimaki, H. Nanoparticles and Neurotoxicity. IJMS 2011, 12, 6267–6280. [Google Scholar] [CrossRef] [PubMed]
  176. Huang, Y.-W.; Cambre, M.; Lee, H.-J. The Toxicity of Nanoparticles Depends on Multiple Molecular and Physicochemical Mechanisms. IJMS 2017, 18, 2702. [Google Scholar] [CrossRef]
  177. Bencsik, A.; Lestaevel, P.; Guseva Canu, I. Nano- and Neurotoxicology: An Emerging Discipline. Prog. Neurobiol. 2018, 160, 45–63. [Google Scholar] [CrossRef] [PubMed]
  178. Jesus, S.; Schmutz, M.; Som, C.; Borchard, G.; Wick, P.; Borges, O. Hazard Assessment of Polymeric Nanobiomaterials for Drug Delivery: What Can We Learn From Literature So Far. Front. Bioeng. Biotechnol. 2019, 7, 261. [Google Scholar] [CrossRef]
  179. Makadia, H.K.; Siegel, S.J. Poly Lactic-Co-Glycolic Acid (PLGA) as Biodegradable Controlled Drug Delivery Carrier. Polymers 2011, 3, 1377–1397. [Google Scholar] [CrossRef]
  180. Shvedova, A.; Pietroiusti, A.; Kagan, V. Nanotoxicology Ten Years Later: Lights and Shadows. Toxicol. Appl. Pharmacol. 2016, 299, 1–2. [Google Scholar] [CrossRef] [PubMed]
  181. Shang, L.; Nienhaus, K.; Nienhaus, G.U. Engineered Nanoparticles Interacting with Cells: Size Matters. J. Nanobiotechnol. 2014, 12, 5. [Google Scholar] [CrossRef] [PubMed]
  182. Yuan, Z.-Y.; Hu, Y.-L.; Gao, J.-Q. Brain Localization and Neurotoxicity Evaluation of Polysorbate 80-Modified Chitosan Nanoparticles in Rats. PLoS ONE 2015, 10, e0134722. [Google Scholar] [CrossRef] [PubMed]
  183. Caliceti, P. Pharmacokinetic and Biodistribution Properties of Poly(Ethylene Glycol)–Protein Conjugates. Adv. Drug Deliv. Rev. 2003, 55, 1261–1277. [Google Scholar] [CrossRef]
  184. Martins, C.; Sousa, F.; Araújo, F.; Sarmento, B. Functionalizing PLGA and PLGA Derivatives for Drug Delivery and Tissue Regeneration Applications. Adv. Healthc. Mater. 2018, 7, 1701035. [Google Scholar] [CrossRef]
  185. Dogan, A.E.; Yuksel, C.; Du, F.; Chouinard, V.-A.; Öngür, D. Brain Lactate and pH in Schizophrenia and Bipolar Disorder: A Systematic Review of Findings from Magnetic Resonance Studies. Neuropsychopharmacology 2018, 43, 1681–1690. [Google Scholar] [CrossRef]
  186. Rabha, B.; Bharadwaj, K.K.; Pati, S.; Choudhury, B.K.; Sarkar, T.; Kari, Z.A.; Edinur, H.A.; Baishya, D.; Atanase, L.I. Development of Polymer-Based Nanoformulations for Glioblastoma Brain Cancer Therapy and Diagnosis: An Update. Polymers 2021, 13, 4114. [Google Scholar] [CrossRef]
  187. Pinto, M.; Silva, V.; Barreiro, S.; Silva, R.; Remião, F.; Borges, F.; Fernandes, C. Brain Drug Delivery and Neurodegenerative Diseases: Polymeric PLGA-Based Nanoparticles as a Forefront Platform. Ageing Res. Rev. 2022, 79, 101658. [Google Scholar] [CrossRef] [PubMed]
  188. Mahmoudi, M.; Bertrand, N.; Zope, H.; Farokhzad, O.C. Emerging Understanding of the Protein Corona at the Nano-Bio Interfaces. Nano Today 2016, 11, 817–832. [Google Scholar] [CrossRef]
  189. Raman, S.; Mahmood, S.; Hilles, A.R.; Javed, M.N.; Azmana, M.; Al-Japairai, K.A.S. Polymeric Nanoparticles for Brain Drug Delivery—A Review. Curr. Drug Metab. 2020, 21, 649–660. [Google Scholar] [CrossRef]
  190. Chen, C.C.; Sheeran, P.S.; Wu, S.-Y.; Olumolade, O.O.; Dayton, P.A.; Konofagou, E.E. Targeted Drug Delivery with Focused Ultrasound-Induced Blood-Brain Barrier Opening Using Acoustically-Activated Nanodroplets. J. Control. Release 2013, 172, 795–804. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of the most diffused conventional plastics and bioplastics.
Figure 1. Chemical structures of the most diffused conventional plastics and bioplastics.
Pharmaceutics 15 02549 g001
Figure 2. Overview of microplastics (MPs) and nanoplastics (NPs) destiny from the environment up to biological effects. BBB, blood–brain barrier.
Figure 2. Overview of microplastics (MPs) and nanoplastics (NPs) destiny from the environment up to biological effects. BBB, blood–brain barrier.
Pharmaceutics 15 02549 g002
Figure 3. A schematic illustration of the principal manufacturing methods to produce polymeric micro/nano-particles: conventional nanoprecipitation by anti-solvent effect (a), nanoprecipitation enhanced by adopting microfluidics channel (b), solvent extraction/evaporation of emulsion (c), supercritical fluids emulsion extraction (d), spray drying (e).
Figure 3. A schematic illustration of the principal manufacturing methods to produce polymeric micro/nano-particles: conventional nanoprecipitation by anti-solvent effect (a), nanoprecipitation enhanced by adopting microfluidics channel (b), solvent extraction/evaporation of emulsion (c), supercritical fluids emulsion extraction (d), spray drying (e).
Pharmaceutics 15 02549 g003
Figure 4. Chemical and physical characterization of the polymeric nanoparticles.
Figure 4. Chemical and physical characterization of the polymeric nanoparticles.
Pharmaceutics 15 02549 g004
Figure 5. Pros and cons of bioplastic MPs/NPs drug delivery systems for brain disease management.
Figure 5. Pros and cons of bioplastic MPs/NPs drug delivery systems for brain disease management.
Pharmaceutics 15 02549 g005
Table 3. Preclinical studies on bioplastic MPs/NPs used as drug delivery systems in brain diseases.
Table 3. Preclinical studies on bioplastic MPs/NPs used as drug delivery systems in brain diseases.
BiopolymerTechnologyMean Size (nm)Drug LoadedIn Vitro/In Vivo ModelDataBrain DiseaseRef.
Chi-PLGANP136 ± 30Lutein-Co-culture model of BBB
-Nasal mucosa
-Male Wistar rats
Oxidative stress reductionAD[173]
PLGASE226.2 ± 40Thymoquinone-Male ratsꜛ SOD activity
ꜜ Oxidative stress
AD[145]
PEG-PLGANP145.2 ± 43Rhynchophylline-bEnd-3 cells
-C57BL/6 mice
-Pass through BBB
-Regulate neuronal activity
AD[147]
PEGNP125 ± 65Functionalized Ab anti Aβ-AD transgenic mice
-male Tg2576
-Correction of memory deficit
-Reduction in Aβ levels
AD[149]
PLGASE497 ± 353Dopamine-6-OHDA PD Swiss mice-Replenishment dopamine level
-Motor symptoms improvement
PD[154]
PLGASE720 ± 87Levodopa-PC-12 neuronal like cells
-CD57/BL6 mice
-Improvement drug delivery
ꜛ Therapeutic efficacy
PD[155]
PEG-lipid-PLCL-NPNP150 ± 50Dopamine/Tenoxicam
Lamotrigine
-Human cortical neuronal cells-2 (HCN2)
-BBB hCMEC/D3 cell line
-Male Wistar rats
Cognitive improvementPD[156]
PEG-PLA-SE106 ± 5.4Adapalene-OD1G93A transgenic mice-Improvement of adapalene encapsulation
-Activation of retinoid signaling pathway
-Improvement motor performance
ALS[162]
PLGASE521 ± 289with MOG-C57BL/6 miceAmeliorates autoimmune responseMS[164]
PLGASE~110Doxorubicina-Male Wistar ratsAntitumor effectsGlioblastoma[167]
PLGASE250 ± 180 Chlorotoxin-conjugated morusin-U87 and GI-1 human glioma cellsAntitumor effectsGlioblastoma[169]
Abbreviations: PLGA, polylactic co-glycolic acid; Chi, chitosan coated; PEG, polyethylene glycol; PLCL, poly (l-lactide-co-ε-caprolactone); NP, nanoparticle; PNP, polymeric nanoparticle; MOG, myelin oligodendrocyte glycoprotein; BBB, blood–brain barrier; SOD, superoxide dismutase; AD, Alzheimer’s disease; PD, Parkinson’s disease; ALS, amyotrophic lateral sclerosis; MS, multiple sclerosis; NP, nanoprecipitation; SE, solvent evaporation. ꜛ increase; ꜜdecrease.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lamparelli, E.P.; Marino, M.; Szychlinska, M.A.; Della Rocca, N.; Ciardulli, M.C.; Scala, P.; D'Auria, R.; Testa, A.; Viggiano, A.; Cappello, F.; et al. The Other Side of Plastics: Bioplastic-Based Nanoparticles for Drug Delivery Systems in the Brain. Pharmaceutics 2023, 15, 2549. https://doi.org/10.3390/pharmaceutics15112549

AMA Style

Lamparelli EP, Marino M, Szychlinska MA, Della Rocca N, Ciardulli MC, Scala P, D'Auria R, Testa A, Viggiano A, Cappello F, et al. The Other Side of Plastics: Bioplastic-Based Nanoparticles for Drug Delivery Systems in the Brain. Pharmaceutics. 2023; 15(11):2549. https://doi.org/10.3390/pharmaceutics15112549

Chicago/Turabian Style

Lamparelli, Erwin Pavel, Marianna Marino, Marta Anna Szychlinska, Natalia Della Rocca, Maria Camilla Ciardulli, Pasqualina Scala, Raffaella D'Auria, Antonino Testa, Andrea Viggiano, Francesco Cappello, and et al. 2023. "The Other Side of Plastics: Bioplastic-Based Nanoparticles for Drug Delivery Systems in the Brain" Pharmaceutics 15, no. 11: 2549. https://doi.org/10.3390/pharmaceutics15112549

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop