Nanoparticles can be used for drug delivery and consist of many sizes and chemical compositions. They can accommodate a diverse population of drugs and can be made to target specific areas of the body. Fabrication methods generally follow either top-down or bottom-up manufacturing techniques, which have differing production controls, which determine nanoparticle characteristics including but not limited to size and encapsulation efficiency. Functionalizing these nanoparticles is done to add drugs, prevent aggregation, add positive charge, add targeting, etc. As the nanoparticles reach the target cells, cellular uptake occurs, drug is released, and the nanoparticle is broken down. Poly(lactic-co-glycolic acid) (PLGA) nanoparticles have often been used for drug delivery applications as they have shown minimal toxicity, which has helped with US FDA approval. This review breaks down PLGA nanoparticle fabrication, functionalization, and biological considerations.

Poly(lactic-co-glycolic acid) (PLGA) nanoparticles have been used as a vessel for drug delivery for some time. Some examples of PLGA nanoparticles are included in Table I. PLGA is an established US FDA approved polymer used in many nanoparticle applications due to its high biocompatibility and biodegradability.1–3 Bare PLGA nanoparticles, however, will not do much inside of the body and so research has been done to functionalize these nanoparticles for various applications. With the newly targeted applications, researchers needed to consider new sizes, shapes, drugs, surface features, and biology, which are largely accompanied by new fabrication methods.

TABLE I.

List of US FDA approved drugs that contain PLGA. Note the approval years, some older and some newer.

Drug nameActive ingredientCompanyApplicationYear approved
Lupron Depot Leuprolide acetate AbbVie Endocrine Inc Advanced Prostatic cancer 1989 
Sandostatin LAR Octreotide acetate Novartis Acromegaly, carcinoid tumors, Vasoactive intestinal peptide Tumors 1998 
Eligard Leuprolide acetate Tolmar Therapy Advanced prostatic cancer 2002 
Risperdal Consta Risperidone Janssen Pharms Schizophrenia, bipolar disorder 2007 
Bydureon Exenatide synthetic AstraZeneca AB Type 2 diabetes Mellitus 2012 
Drug nameActive ingredientCompanyApplicationYear approved
Lupron Depot Leuprolide acetate AbbVie Endocrine Inc Advanced Prostatic cancer 1989 
Sandostatin LAR Octreotide acetate Novartis Acromegaly, carcinoid tumors, Vasoactive intestinal peptide Tumors 1998 
Eligard Leuprolide acetate Tolmar Therapy Advanced prostatic cancer 2002 
Risperdal Consta Risperidone Janssen Pharms Schizophrenia, bipolar disorder 2007 
Bydureon Exenatide synthetic AstraZeneca AB Type 2 diabetes Mellitus 2012 

Two major methods exist to fabricate nanoparticles: emulsification-solvent evaporation and precipitation. Each has its advantages and disadvantages, with emulsification-solvent evaporation being able to be more easily scaled up where precipitation can achieve higher encapsulation efficiencies. Precipitation also includes the popular microfluidic methods that report fine-tuning of nanoparticle size, charge, and encapsulation efficiency. PLGA nanoparticles can be functionalized by choosing what drug is being loaded and by modifying the nanoparticle surface. After the nanoparticles are in the body, researchers need to consider passive targeting, the efficiency of cellular uptake, the release profile of any carried drugs, and the degradation/clearance/toxicity.

This review aims to provide an overview of PLGA nanoparticles starting with their fabrication, then how they are popularly functionalized, and finally what factors are often considered once the nanoparticles interact with the body. A researcher can use the contents of this review to jumpstart their design of a PLGA nanoparticle for a new application.

Top-down manufacturing is a process where an item is made by breaking down larger items like chopping wood. In terms of PLGA nanoparticle synthesis, top-down refers to the breaking down of a bulk precursor to nanoparticles using some mechanical method. As seen in Fig. 1, one common method is emulsification-solvent evaporation, which consists of applying high shear force (i.e., sonication/ultrasonication) to a mixture of a small amount of PLGA dissolved in water-immiscible organic solvent and a large amount of surfactant dissolved in water and then evaporating out the organic solvent.4–10 Many examples of various single emulsion techniques have been explored with varying success.5,8–14 The initial mixture does not form any nanoparticles since the organic solvent is not miscible in water, and so pockets of PLGA in organic solvent float around in the water, unable to form nanoparticles until agitated in some way. Using sonication as the example shear force, once the probe sonicator is introduced, the energy introduced forces the water-immiscible organic solvent to mix with the water, thus exposing PLGA to water. Since PLGA is hydrophobic, it will clump together in water and form nanoparticles. In this sense, the nanoparticles formed have no layer or bilayer and are, instead, simply a collection of PLGA polymers in a ball. The size of the nanoparticles that form initially is large but continues to decrease as the sonication continues. While sonication causes the initial mixing and nanoparticle formation, it also breaks up the nanoparticles after they have been formed to allow them to reform, typically at a smaller size. When taken to a limiting amount of time, nanoparticles will be broken down and reformed to the same size, based on the sonicator strength, and the sonication is complete. The removal of the organic solvent via evaporation reduces the nanoparticle size and is necessary afterward because the organic solvent is harmful to the nanoparticle actions and toxicity. PLGA nanoparticle properties can be detected in numerous ways including but not limited to electron microscopy, Dynamic light scattering (DLS), AFM, and fluorescence detection.15 

FIG. 1.

Emulsification-solvent evaporation single emulsion process. Sonication used as an example method to apply high shear force.

FIG. 1.

Emulsification-solvent evaporation single emulsion process. Sonication used as an example method to apply high shear force.

Close modal

Since this organic solvent is immiscible in water, some examples are triethylamine,16 dichloromethane (DCM),5–8,12–14,16–24 chloroform,9,16,25,26 diethyl ether,16 and ethyl acetate.11,16 Many considerations come into play when determining which organic solvent is right, but sometimes, the most important parameter for fabrication is the length of the evaporation step. Diethyl ether has the shortest evaporation step and so it would be chosen if time was the largest factor.27 The toxicity of the chosen organic solvent may also be a greater factor when going for US FDA approval as substances like chloroform are highly toxic and need to be 100% removed to be deemed safe and getting to 100% removal will inevitably increase either fabrication time or cost.

Described earlier was single emulsion, where hydrophobic drugs would be used, but double emulsion has also been demonstrated, which allows the use of hydrophilic drugs as shown in Fig. 2.6,7,17–26,28 Double emulsion literally means two emulsions and consists of adding a small amount of water to the organic solvent before the organic solvent is added to the bulk water. Hydrophilic drugs are dissolved in the small amount of water added to the organic solvent. This is better than just adding hydrophilic drugs to the bulk water as the encapsulation efficiency is higher since more drug is near the PLGA when the nanoparticles are forming. If you add hydrophilic drugs to the bulk water, then there is a higher chance the drug never reaches any PLGA and, thus, is never encapsulated.

FIG. 2.

Double emulsion process. Sonication again used as an example method to apply high shear force.

FIG. 2.

Double emulsion process. Sonication again used as an example method to apply high shear force.

Close modal

Bottom-up manufacturing refers to the building up of a larger item from smaller pieces like building a house. For PLGA nanoparticle synthesis, bottom-up means forming nanoparticles from single polymer strands of PLGA. This has been done by adding an organic solvent to water just as before in a process called precipitation (nanoprecipitation for nanoparticle formation, except this organic solvent is miscible with water).4,29–40 With light magnetic stirring, the two phases mix readily and form nanoparticles immediately.

The organic phase used here needs to be miscible in water, so some examples are acetone,16,30,31,33,35,37,39,40 acetonitrile,16 dimethyl sulfoxide (DMSO),16 and tetrahydrofuran (THF).16,32 Again, the choice would be based on several factors including evaporation time and toxicity.

To manufacture a significant amount of nanoparticles for clinical and commercial applications, the scale-up of the process is necessary. These methods need to overcome several obstacles including reproducibility, toxicity, and surface chemistry.41,42 Existing solutions use membrane extrusion,43,44 supercritical fluid technology,45 and spinning disk processing46 each with its own advantages and disadvantages.

An adaptation to the previously mentioned bulk methods is the use of microfluidics and hydrodynamic flow focusing (HFF).47–52 In the HFF technique, the two phases are prepared the same but are then injected into a micro-/nano-scale polydimethylsiloxane (PDMS) device, which is manufactured by a combination of UV lithography and PDMS molding steps, where the two phases meet. In this type of device, the organic phase is pinched on both sides by the water phase. As soon as the phases meet, diffusion begins resulting in nanoparticle formation. To control the size and encapsulation efficiency, one can adjust several parameters including each phase's flow rate, the flow rate ratio, the channel length, all concentrations, and the materials are used. An additional material consideration with microfluidic methods is the interaction between PDMS and PLGA. Since normal PDMS is hydrophobic, it attracts PLGA and, by extension, the PLGA nanoparticles, leading to aggregation in the channels and eventually clogging. Methods have been developed to prevent this aggregation/clogging such as using oxygen plasma to oxidize the surface of the channels, which makes the surface hydrophilic and, thus, repels PLGA and the formed nanoparticles. Another challenge with microfluidics is the scale-up of the process as the working volume, though advantageous for size control and encapsulation efficiency, also limits the throughput.53,54 For microfluidic devices, scale-up is necessary to increase the naturally low throughput and can be achieved by employing parallelization and switching from PDMS to another material as PDMS cannot handle the higher flow rates required for increased throughput.55–57 Since many microfluidic scale-ups require making a more complex device, increases in the manufacturing cost and/or time can be expected.

After nanoparticles are produced by any method, they need to be stored properly to maintain their stability. The method (i.e., aqueous, freezing, and lyophilized storage) and type of cryoprotectant (i.e., sucrose, trehalose, or mannitol) greatly affect stability with notably the only US FDA approved COVID-19 mRNA vaccines, which are made from lipid nanoparticles, being stored in freezing conditions with sucrose.4,58,59

Both fabrication methods can benefit greatly from various PLGA nanoparticle manipulation techniques. Bulk methods tend to employ ultracentrifugation,15,60,61 ultrafiltration,15,62 size exclusion chromatography,15,63 precipitation,15,64 or immunoaffinity techniques,15,65 which are used to separate, purify, and concentrate the PLGA nanoparticles. Microfluidic methods can employ either active or passive techniques. Active techniques include acoustic,15,66 electric,15,67 magnetic,15,68 and optical controls,15,69 whereas passive techniques include inertial microfluidics,15,70 deterministic lateral displacement,15,71 microfluidic filtration,15,72 pinched flow fractionation,15,73 and viscoelastic microfluidics.15,74 Both active and passive techniques aim to control particles with active methods tending to be more precise but having lower throughput and passive methods tending to be less precise but having higher throughput.15,75

Nanoparticles can hold and deliver many different types of drugs and other substances including but not limited to doxorubicin (cancer),19 paclitaxel (cancer),9,12,39,76 Vascular endothelial growth factor (VEGF) (regenerative medicine),77 and various nucleic acids (whatever they code for, numerous applications).21,26,39,59,78 Drug/substance choice plays a major role in the determining fabrication method and based on whether the drug/substance is soluble in water or not. Doxorubicin and nucleic acids, for example, are water-soluble, whereas paclitaxel and VEGF are not.

The water-soluble drugs and substances must be dissolved in water, so for the emulsification-solvent evaporation method, double emulsion must be used.19,21,26,39,59,78 Water-soluble drugs are, thus, in the first water phase before adding to the organic solvent phase and then the bulk water phase. If you dissolve the drug in the bulk water phase, the encapsulation efficiency will be very low as most drug will never see any PLGA, so there is a lowered chance the drug is encapsulated whereas if the drug is dissolved in the first water phase, then all the drug has the potential to see PLGA as it is all added to the organic solvent phase first. For the precipitation method, since the organic solvent and water dissolve readily, it is important to keep the volume lower to increase the encapsulation efficiency. The larger the working volume, the higher chance the drug never sees PLGA, and increasing the amount of organic solvent phase creates a new problem as more nanoparticles will then end up forming with no drug, which is harder to separate out at the end as they should all roughly be the same size. Therefore, a technique like the microfluidic HFF, as shown in Fig. 3, will lead to higher encapsulation efficiency since the working volume is much smaller than bulk mixing and the ratio of organic solvent to water can be carefully managed.

FIG. 3.

Microfluidic 3DHFF nanoparticle fabrication steps. shRNA is dissolved in the outer aqueous phase and mixed with the organic phase where the channels meet. The water and acetonitrile go through solvent exchange on the barrier of the phases. After solvent exchange, some PLGA is now in a water-rich environment and so it self-assembles into a nanoparticle. Solvent exchange continues over the length of the channel wherever the aqueous phase meets the organic phase with the nanoparticles growing over the length of the channel. Adjusting the speeds of the channels and the ratio of speeds can tune the final size of the nanoparticles.

FIG. 3.

Microfluidic 3DHFF nanoparticle fabrication steps. shRNA is dissolved in the outer aqueous phase and mixed with the organic phase where the channels meet. The water and acetonitrile go through solvent exchange on the barrier of the phases. After solvent exchange, some PLGA is now in a water-rich environment and so it self-assembles into a nanoparticle. Solvent exchange continues over the length of the channel wherever the aqueous phase meets the organic phase with the nanoparticles growing over the length of the channel. Adjusting the speeds of the channels and the ratio of speeds can tune the final size of the nanoparticles.

Close modal

Drugs like paclitaxel and the cytokine VEGF on the other hand cannot be dissolved in water and, therefore, are dissolved in the organic solvent.9,12,39,76,77 This means that the simpler single emulsion technique can be used for the emulsification-solvent evaporation method. Water-insoluble drugs are in the organic solvent phase, which is added to the bulk water phase, so all the drug has a chance to be encapsulated. Since the drugs are dissolved in the organic phase, simple bulk nanoprecipitation can be used for the precipitation method. If the microfluidic HFF technique is desired, the organic solvent phase should be switched with the water phase, so the organic solvent phase is in the middle channel and is squeezed by water phases on both sides. This would keep the drug, which is in the organic solvent phase, in the center of the channel and, thus, able to see PLGA. If you kept the organic solvent phase on the outside, then the formed nanoparticles on the water boundary would be more likely to contain no drug. The microfluidic method is still attractive compared to the bulk method due to its better experimental control, smaller working volume, and finer parameter tuning.

PLGA nanoparticles themselves are simply a ball of PLGA and so they do not necessarily have control of where they are delivered, their surface charge, and the potential for collisions with other PLGA nanoparticles, resulting in aggregations or increased size. PLGA nanoparticles can have their surface modified with the addition of many different types of molecules including various targeting ligands, chitosan, lipids, polyethylene glycol (PEG), and surfactants as listed in Table II.

TABLE II.

List of surface modifications. Additional secondary functions and examples in the text.

Surface modificationMain function(s)Example(s)Reference(s)
Ligands Cell targeting Antibodies 9, 79–110  
Biotin 
Bisphosphonates 
Folic acid 
Lectins 
Mannan 
Aptamers 
Peptides 
Sialic acid 
Transferrin 
Chitosan Adds positive charge to NP Improves cellular uptake Chitosan 6, 14, 20, 31, 111  
Lipids Increase encapsulation efficiency Phospholipid 59, 78, 112–118  
Assist cellular uptake Cationic lipid 
Encourage endosomal escape Anionic lipid 
Can be charged Cholesterol 
PEG NP shielding PEG 8–10, 32, 35, 36, 78, 119  
Surfactants NP stabilization PVA 5–9, 11–13, 17–19, 21–23, 26, 28–30, 34–36, 40, 120  
Can be charged Pluronics 
Polysorbates 
Poloxamers 
DMAB 
Surface modificationMain function(s)Example(s)Reference(s)
Ligands Cell targeting Antibodies 9, 79–110  
Biotin 
Bisphosphonates 
Folic acid 
Lectins 
Mannan 
Aptamers 
Peptides 
Sialic acid 
Transferrin 
Chitosan Adds positive charge to NP Improves cellular uptake Chitosan 6, 14, 20, 31, 111  
Lipids Increase encapsulation efficiency Phospholipid 59, 78, 112–118  
Assist cellular uptake Cationic lipid 
Encourage endosomal escape Anionic lipid 
Can be charged Cholesterol 
PEG NP shielding PEG 8–10, 32, 35, 36, 78, 119  
Surfactants NP stabilization PVA 5–9, 11–13, 17–19, 21–23, 26, 28–30, 34–36, 40, 120  
Can be charged Pluronics 
Polysorbates 
Poloxamers 
DMAB 

For PLGA nanoparticles to be effective, they must be delivered to the right cells in the body. Adding cell-targeting ligands via physical association or chemical conjugation allows the PLGA nanoparticle to be internalized by receptor-mediated endocytosis after ligand–receptor binding. Ligands can be added by either electrostatic interactions where a positively charged group attached to the ligand is attracted to the negatively charged PLGA surface or through hydrophobic interactions where a ligand attached to a hydrophobic chain is attracted to the hydrophobic PLGA surface.79 Chemically, a ligand can be attached by carbodiimide (carboxyl–amine reaction), maleimide (maleimide–thiol reaction), or click chemistry (alkyne–azide reaction).121,122 The most common ligands are antibodies that only interact with their specific surface antigen including anti-CD133 (gastric carcinoma cells),9,79,80 anti-Prostate-specific membrane antigen (PSMA) (prostate cancer cells),79,81 and anti-HER2 (ovarian and human breast cancer cells).79,82–84 Other ligands include biotin (cancer),79,85 bisphosphonates (bone-related conditions),79,86–88 folic acid (brain, breast, cervical, colorectal, epithelial, kidney, lung, and ovarian tumors),79,89 lectins (cancer),79,90–93 mannan (macrophages and dendritic cells),79,94 aptamers (designed against any cancer-related biomarker),79,95–98 peptides (cancer and atherosclerotic plaques),79,99–108 sialic acid (cancer, leukocytes, platelets, and endothelial cells),79,109 and transferrin (breast cancer, pancreatic cancer, lung cancer, and brain gliomas).79,110

Chitosan is a polycationic polymer made from chitin that has been used for numerous biomedical applications including being added on to existing nanoparticles and serving as the base polymer for nanoparticles. For PLGA nanoparticles, it is used to add a positive charge to the surface of the nanoparticles.6,14,20,31,111 A positive surface charge on nanoparticles allows the nanoparticle to uptake into the cell more readily as charged nanoparticles (positive and negative) show preferential uptake in cells with positive charged nanoparticles showing the highest.111,123,124 Chitosan can be added to the surface of PLGA nanoparticles through simple bulk mixing as the PLGA gives the nanoparticle a negative charge, which will attract the positively charged chitosan. Since chitosan is positively charged, any negatively charged drug that is in the bulk when it is added will be attracted to it. This could result in a negatively charged drug being entrapped onto the surface of the nanoparticle, as opposed to within the ball of PLGA. The release profile could then be negatively impacted and result in incorrect encapsulation efficiency reporting as the drug on the surface would be counted as the entrapped drug. This issue can be avoided by adding chitosan after nanoparticle formation so that no drug is in the bulk phase when the chitosan is added.

Many types of lipids have been added to PLGA nanoparticles to enable monodisperse fabrication, improve stability, increase encapsulation efficiency of nucleic acids, assist cellular uptake, and encourage endosomal escape of nucleic acids.59,78,112–118 Phospholipids are types of lipids that are comprised of a hydrophilic “head” group containing a phosphate group, two hydrophobic “tails” that are derived from fatty acids, and a glycerol molecule to link them together. This amphiphilic nature causes phospholipids to self-arrange into a layer when in a solution containing water and organic solvent, as is the case when fabricating PLGA nanoparticles. The hydrophilic head will orient toward the water with the hydrophobic tails orienting toward the organic solvent. When surrounding a droplet of organic solvent, these phospholipids will self-arrange into a monolayer around the organic solvent forming a micelle. This will surround anything in that organic solvent droplet, which is the ball of PLGA for PLGA nanoparticles. The incorporation of phospholipids, therefore, increases stability and circulation time. The addition of cationic lipids has been shown to increase the encapsulation efficiency of nucleic acids due to the positive charge on the lipid attracting the negatively charged nucleic acids.59,78,113,117,118 Adding cholesterol maintains membrane integrity by matching the molar ratio to endogenous membranes as well as decreases the number of surface-bound proteins and improves circulation time.

PEG is a polyether compound that is commonly added to PLGA nanoparticle surfaces to help the nanoparticle avoid the mononuclear phagocytic system (functions as a shielding group). In short, this allows the PLGA nanoparticle to avoid phagocytosis and, thus, reach the intended target location. PEG is a common choice as a shielding group as it is electrically neutral, has high hydrophilicity, and shows high spatial repulsion.8–10,32,35,36,78,119 PEG can be added via physical absorption (as part of a hydrophobic or charged group), covalent coupling (as part of a reactive group), or self-assembly (as part of a hydrophobic copolymer including lipids).119 PEG has also been shown to improve the nanoparticle stability through steric repulsion, decrease the encapsulation efficiency of hydrophilic drugs, increase the encapsulation efficiency of hydrophobic drugs, and be susceptible to the accelerated blood clearance, which is a phenomenon where the circulating half-life of PEGylated nanoparticles is shortened after a second injection of nanoparticles is introduced, theorized to be the result of the spleen producing and releasing anti-PEG antibodies after the first injection.59,78,112–117,119,125

Surfactants are compounds that decrease surface tension between two materials, acting as a stabilizing compound. Examples include various pluronics,8,30,40,120 polysorbates,36,120 and poloxamers,29,35,36,120 as well as the commonly used polyvinyl alcohol (PVA).5–7,9,12,13,17–19,21–23,26,28,34,40,120 For PLGA nanoparticles, a surfactant is added to seal the nanoparticles, so they do not grow or aggregate. They consist of a hydrophilic head and a hydrophobic tail so when they interact with a nanoparticle, the tails orient toward the PLGA, and the heads orient toward the water. This results in the creation of a monolayer around the ball of PLGA. They can be further classified as nonionic and ionic depending on the charge of the head. Different surfactants also maintain different sizes of nanoparticles by changing the contact angle. Surfactants can also play a role in delivery as they can allow the surrounded PLGA core to avoid phagocytosis when attempting to reach the target area. The surfactant didodecyldimethylammonium bromide (DMAB) is unique as it has an inherent positive charge, thus removing the need for chitosan or other additions that add positive charge to the nanoparticle.11 

When PLGA nanoparticles enter the body, there are several major factors to consider. These being passive targeting, efficiency of cellular uptake, the release profile of any carried drugs, and the degradation/clearance/toxicity of the nanoparticle as seen in Table III. Accounting for all these factors will allow any nanoparticle to perform its function while not harming the body in any way.

TABLE III.

List of biological considerations. Additional secondary factors and examples in text.

Biological considerationsMain factor(s)Affect(s)
Passive targeting Size Where in the body NP uptake occurs 
Cellular uptake Size Whether or not cellular uptake occurs 
Shape Rate pathway of cellular uptake 
Surface modifications 
Release profile Attached vs entrapped drug size Drug release pathway and timing 
Degradation and clearance Shape Circulation time, toxicity, and environmental effects 
Surface modifications 
Biological considerationsMain factor(s)Affect(s)
Passive targeting Size Where in the body NP uptake occurs 
Cellular uptake Size Whether or not cellular uptake occurs 
Shape Rate pathway of cellular uptake 
Surface modifications 
Release profile Attached vs entrapped drug size Drug release pathway and timing 
Degradation and clearance Shape Circulation time, toxicity, and environmental effects 
Surface modifications 

Targeting can be controlled by adding surface modifications as mentioned above (active targeting), but targeting can also be done by controlling the size of the PLGA nanoparticle (passive targeting). There are cases where active targeting is undesirable such as to avoid creating a binding site barrier effect and to reduce toxicity.126–128 In these cases, size control becomes very important in determining where the nanoparticles will uptake. Generally, nanoparticles must be within the 10–200 nm range as nanoparticles less than 10 nm in diameter are rapidly eliminated by the kidneys and nanoparticles greater than 200 nm in diameter cause inflammation and phagocytic activity.129–132 Examples for specific cells are that sizes of 30–50 nm are desired when targeting pancreatic cancer cells, sizes of 40–60 nm are better for targeting breast cancer cells, sizes around 100 nm are optimal for targeting the gastrointestinal tract, and sizes around 120 for respiratory epithelium.129,133–135

Cellular uptake of PLGA nanoparticles can occur in a few ways depending on the size, shape, and surface modifications (including charge). Five common pathways for cellular uptake are phagocytosis, caveolin-mediated endocytosis, clathrin-mediated endocytosis, independent endocytosis, and macropinocytosis each being more likely for certain nanoparticle conformations.129,136 Phagocytosis of nanoparticles occurs for larger particles like those greater than 200 nm and is initiated by opsonization (adsorption of antibodies onto the nanoparticle surface), which is then recognized by phagocytes and internalized.132,137 Surface modification by adding PEG can act as a shield, essentially preventing opsonization and, therefore, phagocytosis.137 Caveolin-mediated endocytosis happens where caveolin is located on the cell membrane. Uptake forms a 50–80 nm size vesicle, which can escape lysosomal degradation.138 Clathrin-mediated endocytosis takes place where clathrin is located on the plasma membrane and creates vesicles with diameters of 100–150 nm but are more susceptible to lysosomal degradation.139,140

Independent endocytosis occurs in cells without clathrin and caveolae, and an example is folate-modified nanoparticles, which also might escape trafficking into lysosomes.141,142 Macropinocytosis is for large nanoparticles that are too big for caveolae- or clathrin-mediated endocytosis and consists of bulk fluid uptake of all particles and dissolved molecules in the extracellular fluid.143 Nanoparticle shape affects uptake at certain sizes. For nanoparticles greater than 100 nm in diameter, the order of highest uptake to lowest was rods, spheres, cylinders, and then cubes.144 For nanoparticles less than 100 nm in diameter, spheres showed the highest uptake.145,146 Using needle-shaped PLGA nanoparticles was attempted and showed increased uptake but with the increased risk of lysosome disruption leading to cell apoptosis.147 Although the size dictates the pathway, MacCuaig et al. have shown that active targeting (surface modification) has a much larger impact on uptake than passive targeting (size/shape control).134 As mentioned above, adding certain surface modifications improves cellular uptake like the addition of antibodies or peptides when targeting cancers or certain lipids when targeting specific areas of the body.134,148 Also from above, positively charged nanoparticles (through surface addition of chitosan) show preferential uptake as the cell membrane possesses a slight negative charge and, thus, there are some electrostatic attractions influencing cellular uptake.123,124,149,150

PLGA nanoparticles for delivery must consider the release profile after uptake into a cell. The desired speed at which the delivered drug is released is disease-dependent and so the nanoparticle should be tailored to each case. Generally for PLGA nanoparticles, the burst release of the drug is followed by a prolonged release as there is drug adsorbed on the surface, which releases quickly, and drug entrapped inside the core, which releases slowly.151–156 Smaller nanoparticles intuitively degrade faster and, thus, deliver their drug faster.157,158

After the PLGA nanoparticle delivers its drug, it must then degrade or otherwise clear from the body to limit its toxicity. PLGA can be rapidly cleared by the mononuclear phagocyte system (MPS) as PLGA nanoparticles can diffuse across the permeable vasculature in this system.159 Complete removal of PLGA nanoparticles has been shown to occur within 10 min of administration.160,161 Based on this, if you are targeting rapid release to a location that does not take too long to reach, then this time constraint is of no concern. If you are targeting a location that takes more time, then you will need to employ techniques, some described above, to avoid this rapid clearance. Another way to avoid the MPS while also improving circulation time is to incorporate a hydrophilic shell to your nanoparticle, although the trade-off is increased risk of aggregation and liposome destruction.119 Non-spherical nanoparticles show reduced immune clearance and improved blood circulation time compared to spherical nanoparticles.162,163 The use of certain lipids or PEG can activate the host immune response or be cleared quickly after first administration due to the production of anti-PEG antibodies in a phenomenon known as the accelerated blood clearance effect.117,119,164,165 To avoid this, biodegradable lipids can be used.166–170 Nanoparticles with charged lipids can avoid clearance by the MPS but are susceptible to aggregation in solutions of high ionic strength like blood, which can result in rapid removal from the blood stream.171,172 Nanoparticles made with stabilizing agents such as chitosan, PVA, and Poloxamer 188 showed limited toxicity.173 Additions of nonionic surfactants like Poloxamer 188 also embrace the stealth effect by which opsonization and, thus, phagocytosis is avoided.120 

The use and disposal of PLGA nanoparticles inevitably releases some of them into the environment, leading to potentially negative effects if in a high enough concentration.174,175 They can enter the environment by photochemical transformation, oxidation and reduction, dissolution and precipitation, adsorption and desorption, combustion, biotransformation, and abrasion.176 Material flow analysis and environmental fate modeling detail when PLGA nanoparticle emissions are produced, where they are ultimately deposited, and what happens to them once they are released to the environment.175,177,178 Negative effects of environmental release include nuclei damage and oxidative stress in microorganisms, DNA folding, chromosomal aberrations, and oxidative stress in plants, liver inflammation, reproductive toxicity, and oxidative stress in animals, and shape-based toxicity and oxidative stress in humans.179–186 

PLGA nanoparticle applications grow constantly due to its flexibility in adapting the original bare PLGA nanoparticle by considering new drugs, surface modifications, and biology, as well as the fabrication methods to make those adaptations possible. Continued efforts to expand the field to treat numerous diseases by delivering many new drugs maintain the relevance of research in this field.

The use of nucleic acids as the loaded drug opens the applications of these nanoparticles to almost anything as the design of the nucleic acid can target almost anything. Some challenges remain like the ability to load drugs efficiently and evenly into PLGA nanoparticles with high throughput, the potential to scale up the high throughput processes like microfluidics that have been shown to raise the encapsulation efficiency ceiling, and the means to properly target the nanoparticles so they only go to the target region and nowhere else. As researchers look toward these and other new applications, PLGA nanoparticle fabrication, functionalization, and biological considerations are necessary starting points.

This study was supported by grants from the National Institutes of Health under Grant Nos. R01HL151498 (P. R.K. and B. A.D.) and R41AI149954 (B.A.D., Hilliard Kutscher, and Jonathan F. Lovell).

The authors have no conflicts to disclose.

Eric K. Marecki: Conceptualization (equal); Writing – original draft (equal); Writing – review & editing (equal). Kwang W. Oh: Supervision (equal); Writing – review & editing (equal). Paul R. Knight: Supervision (equal); Writing – review & editing (equal). Bruce A. Davidson: Supervision (equal); Writing – review & editing (equal).

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

1.
Y.
Wang
,
B.
Qin
,
G.
Xia
, and
S. H.
Choi
, “
FDA’s poly(lactic-co-glycolic acid) research program and regulatory outcomes
,”
AAPS J.
23
(
4
),
92
(
2021
).
2.
J. M.
et al, “
Current advances in research and clinical applications of PLGA-based nanotechnology
,”
Expert Rev. Mol. Diagn.
9
(
4
),
325
341
(
2009
) (in English).
3.
M.
Alvi
,
A.
Yaqoob
,
K.
Rehman
,
S. M.
Shoaib
, and
M. S. H.
Akash
, “
PLGA-based nanoparticles for the treatment of cancer: Current strategies and perspectives
,”
AAPS Open
8
(
1
),
12
(
2022
).
4.
K. Y.
Hernández-Giottonini
et al, “
PLGA nanoparticle preparations by emulsification and nanoprecipitation techniques: Effects of formulation parameters
,”
RSC Adv.
10
(
8
),
4218
4231
(
2020
).
5.
C. T. S.
Turk
,
U. C.
Oz
,
T. M.
Serim
, and
C.
Hascicek
, “
Formulation and optimization of nonionic surfactants emulsified nimesulide-loaded PLGA-based nanoparticles by design of experiments
,”
AAPS PharmSciTech.
15
(
1
),
161
176
(
2014
).
6.
M.
Wang
et al, “
Preparation, characterization, and in vitro and in vivo investigation of chitosan-coated poly (d,l-lactide-co-glycolide) nanoparticles for intestinal delivery of exendin-4
,”
Int. J. Nanomed.
8
,
1141
1154
(
2013
) (in English).
7.
M.
Chaturvedi
,
Y.
Molino
,
B.
Sreedhar
,
M.
Khrestchatisky
, and
L.
Kaczmarek
, “
Tissue inhibitor of matrix metalloproteinases-1 loaded poly(lactic-co-glycolic acid) nanoparticles for delivery across the blood-brain barrier
,”
Int. J. Nanomed.
9
,
575
588
(
2014
) (in English).
8.
P.
Yin
et al, “
Bufalin-loaded mPEG-PLGA-PLL-cRGD nanoparticles: Preparation, cellular uptake, tissue distribution, and anticancer activity
,”
Int. J. Nanomed.
7
,
3961
3969
(
2012
) (in English).
9.
S. K.
Swaminathan
,
E.
Roger
,
U.
Toti
,
L.
Niu
,
J. R.
Ohlfest
, and
J.
Panyam
, “
CD133-targeted paclitaxel delivery inhibits local tumor recurrence in a mouse model of breast cancer
,”
J. Controlled Release
171
(
3
),
280
287
(
2013
).
10.
P.
Liu
,
Y.
Sun
,
Q.
Wang
,
Y.
Sun
,
H.
Li
, and
Y.
Duan
, “
Intracellular trafficking and cellular uptake mechanism of mPEG-PLGA-PLL and mPEG-PLGA-PLL-Gal nanoparticles for targeted delivery to hepatomas
,”
Biomaterials
35
(
2
),
760
770
(
2014
).
11.
A.
Xu
et al, “
A physical model for the size-dependent cellular uptake of nanoparticles modified with cationic surfactants
,”
Int. J. Nanomed.
7
,
3547
3554
(
2012
) (in English).
12.
C.
Jin
,
L.
Bai
,
H.
Wu
,
W.
Song
,
G.
Guo
, and
K.
Dou
, “
Cytotoxicity of paclitaxel incorporated in PLGA nanoparticles on hypoxic human tumor cells
,”
Pharm. Res.
26
(
7
),
1776
1784
(
2009
).
13.
R. M.
Mainardes
and
R. C.
Evangelista
, “
PLGA nanoparticles containing praziquantel: Effect of formulation variables on size distribution
,”
Int. J. Pharm.
290
(
1
),
137
144
(
2005
).
14.
T.-W.
Chung
,
S.-S.
Wang
, and
W.-J.
Tsai
, “
Accelerating thrombolysis with chitosan-coated plasminogen activators encapsulated in poly-(lactide-co-glycolide) (PLGA) nanoparticles
,”
Biomaterials
29
(
2
),
228
237
(
2008
).
15.
S.
Hettiarachchi
et al, “
Recent microfluidic advances in submicron to nanoparticle manipulation and separation
,”
Lab Chip
23
(
5
),
982
1010
(
2023
).
16.
See https://organicchemistrydata.org/solvents/ for “Common solvents used in organic chemistry: table of properties,” ACS Division of Organic Chemistry (accessed 2023).
17.
S. J.
Fairley
et al, “
Chlamydia trachomatis recombinant MOMP encapsulated in PLGA nanoparticles triggers primarily T helper 1 cellular and antibody immune responses in mice: A desirable candidate nanovaccine
,”
Int. J. Nanomed.
8
,
2085
2099
(
2013
) (in English).
18.
N.
Samadi
et al, “
The effect of lauryl capping group on protein release and degradation of poly(d,l-lactic-co-glycolic acid) particles
,”
J. Controlled Release
172
(
2
),
436
443
(
2013
).
19.
F.
Li
,
J.
Sun
,
H.
Zhu
,
X.
Wen
,
C.
Lin
, and
D.
Shi
, “
Preparation and characterization novel polymer-coated magnetic nanoparticles as carriers for doxorubicin
,”
Colloids Surf., B
88
(
1
),
58
62
(
2011
).
20.
Y.
Wang
,
P.
Li
, and
L.
Kong
, “
Chitosan-modified PLGA nanoparticles with versatile surface for improved drug delivery
,”
AAPS PharmSciTech
14
(
2
),
585
592
(
2013
).
21.
D.
Cun
,
C.
Foged
,
M.
Yang
,
S.
Frøkjær
, and
H. M.
Nielsen
, “
Preparation and characterization of poly(dl-lactide-co-glycolide) nanoparticles for siRNA delivery
,”
Int. J. Pharm.
390
(
1
),
70
75
(
2010
).
22.
M.
Zaric
et al, “
Skin dendritic cell targeting via microneedle arrays laden with antigen-encapsulated poly-d,l-lactide-co-glycolide nanoparticles induces efficient antitumor and antiviral immune responses
,”
ACS Nano
7
(
3
),
2042
2055
(
2013
).
23.
S.
Sunoqrot
,
J. W.
Bae
,
S.-E.
Jin
,
R. M.
Pearson
,
Y.
Liu
, and
S.
Hong
, “
Kinetically controlled cellular interactions of polymer−polymer and polymer−liposome nanohybrid systems
,”
Bioconjugate Chem.
22
(
3
),
466
474
(
2011
).
24.
S.
Ribeiro
,
N.
Hussain
, and
A. T.
Florence
, “
Release of DNA from dendriplexes encapsulated in PLGA nanoparticles
,”
Int. J. Pharm.
298
(
2
),
354
360
(
2005
).
25.
N.
Rescignano
et al, “
Protein encapsulation in biodegradable polymeric nanoparticles: Morphology, fluorescence behaviour and stem cell uptake
,”
Macromol. Biosci.
13
(
9
),
1204
1212
(
2013
).
26.
H.
Cohen
et al, “
Sustained delivery and expression of DNA encapsulated in polymeric nanoparticles
,”
Gene Ther.
7
(
22
),
1896
1905
(
2000
).
27.
See http://www.stenutz.eu/chem/evaporation.php?s=3 for “Evaporation rates” (accessed 2023).
28.
C.
Colonna
,
R.
Dorati
,
B.
Conti
,
P.
Caliceti
, and
I.
Genta
, “
Sub-unit vaccine against S. aureus-mediated infections: Set-up of nano-sized polymeric adjuvant
,”
Int. J. Pharm.
452
(
1
),
390
401
(
2013
).
29.
C. P.
Reis
,
N.
Martinho
,
C.
Rosado
,
A. S.
Fernandes
, and
A.
Roberto
, “
Design of polymeric nanoparticles and its applications as drug delivery systems for acne treatment
,”
Drug Dev. Ind. Pharm.
40
(
3
),
409
417
(
2014
).
30.
A.
Paul
,
S.
Das
,
J.
Das
,
A.
Samadder
, and
A. R.
Khuda-Bukhsh
, “
Cytotoxicity and apoptotic signalling cascade induced by chelidonine-loaded PLGA nanoparticles in HepG2 cells in vitro and bioavailability of nano-chelidonine in mice in vivo
,”
Toxicol. Lett.
222
(
1
),
10
22
(
2013
).
31.
Z. H.
Wang
,
Z. Y.
Wang
,
C. S.
Sun
,
C. Y.
Wang
,
T. Y.
Jiang
, and
S. L.
Wang
, “
Trimethylated chitosan-conjugated PLGA nanoparticles for the delivery of drugs to the brain
,”
Biomaterials
31
(
5
),
908
915
(
2010
).
32.
H.
Shen
,
X.
Hu
,
M.
Szymusiak
,
Z. J.
Wang
, and
Y.
Liu
, “
Orally administered nanocurcumin to attenuate morphine tolerance: Comparison between negatively charged PLGA and partially and fully PEGylated nanoparticles
,”
Mol. Pharm.
10
(
12
),
4546
4551
(
2013
).
33.
W.
Tao
et al, “
Docetaxel-loaded nanoparticles based on star-shaped mannitol-core PLGA-TPGS diblock copolymer for breast cancer therapy
,”
Acta Biomater.
9
(
11
),
8910
8920
(
2013
).
34.
H.-Y.
Kwon
,
J.-Y.
Lee
,
S.-W.
Choi
,
Y.
Jang
, and
J.-H.
Kim
, “
Preparation of PLGA nanoparticles containing estrogen by emulsification–diffusion method
,”
Colloids Surf., A
182
(
1
),
123
130
(
2001
).
35.
E.
Vega
,
M. A.
Egea
,
A. C.
Calpena
,
M.
Espina
, and
M. L.
García
, “
Role of hydroxypropyl-β-cyclodextrin on freeze-dried and gamma-irradiated PLGA and PLGA-PEG diblock copolymer nanospheres for ophthalmic flurbiprofen delivery
,”
Int. J. Nanomed.
7
,
1357
1371
(
2012
) (in English).
36.
Y.-C.
Chen
,
W.-Y.
Hsieh
,
W.-F.
Lee
, and
D.-T.
Zeng
, “
Effects of surface modification of PLGA-PEG-PLGA nanoparticles on loperamide delivery efficiency across the blood–brain barrier
,”
J. Biomater. Appl.
27
(
7
),
909
922
(
2011
).
37.
S. S.
Bhattacharyya
et al, “
Poly (lactide-co-glycolide) acid nanoencapsulation of a synthetic coumarin: Cytotoxicity and bio-distribution in mice, in cancer cell line and interaction with calf thymus DNA as target
,”
Toxicol. Appl. Pharmacol.
253
(
3
),
270
281
(
2011
).
38.
H.-J.
Jeon
,
Y.-I.
Jeong
,
M.-K.
Jang
,
Y.-H.
Park
, and
J.-W.
Nah
, “
Effect of solvent on the preparation of surfactant-free poly(dl-lactide-co-glycolide) nanoparticles and norfloxacin release characteristics
,”
Int. J. Pharm.
207
(
1
),
99
108
(
2000
).
39.
W. P.
Su
,
F. Y.
Cheng
,
D. B.
Shieh
,
C. S.
Yeh
, and
W. C.
Su
, “
PLGA nanoparticles codeliver paclitaxel and Stat3 siRNA to overcome cellular resistance in lung cancer cells
,”
Int. J. Nanomed.
7
,
4269
4283
(
2012
) (in English).
40.
A. M.
Paiva
et al, “
Development of noncytotoxic PLGA nanoparticles to improve the effect of a new inhibitor of p53–MDM2 interaction
,”
Int. J. Pharm.
454
(
1
),
394
402
(
2013
).
41.
Y.
Herdiana
,
N.
Wathoni
,
S.
Shamsuddin
, and
M.
Muchtaridi
, “
Scale-up polymeric-based nanoparticles drug delivery systems: Development and challenges
,”
OpenNano
7
,
100048
(
2022
).
42.
X.
Liu
and
H.
Meng
, “
Consideration for the scale-up manufacture of nanotherapeutics—A critical step for technology transfer
,”
View
2
(
5
),
20200190
(
2021
).
43.
P.
Guo
,
J.
Huang
,
Y.
Zhao
,
C. R.
Martin
,
R. N.
Zare
, and
M. A.
Moses
, “
Nanomaterial preparation by extrusion through nanoporous membranes
,”
Small
14
(
18
),
1703493
(
2018
).
44.
W.
Xu
et al, “
Generation of polymer nanocapsules via a membrane-extrusion emulsification approach
,”
Mater. Lett.
77
,
96
99
(
2012
).
45.
P.
Pathak
,
M. J.
Meziani
,
T.
Desai
, and
Y.-P.
Sun
, “
Formation and stabilization of ibuprofen nanoparticles in supercritical fluid processing
,”
J. Supercrit. Fluids
37
(
3
),
279
286
(
2006
).
46.
V.
Hakke
,
S.
Sonawane
,
S.
Anandan
,
S.
Sonawane
, and
M.
Ashokkumar
, “
Process intensification approach using microreactors for synthesizing nanomaterials—A critical review
,”
Nanomaterials
11
(
1
),
98
(
2021
).
47.
M.
Ghasemi Toudeshkchouei
,
P.
Zahedi
, and
A.
Shavandi
, “
Microfluidic-assisted preparation of 5-fluorouracil-loaded PLGA nanoparticles as a potential system for colorectal cancer therapy
,”
Materials
13
(
7
),
1483
(
2020
).
48.
A.
Fabozzi
et al, “
Design of functional nanoparticles by microfluidic platforms as advanced drug delivery systems for cancer therapy
,”
Lab Chip
23
(
5
),
1389
1409
(
2023
).
49.
N.
Lababidi
,
V.
Sigal
,
A.
Koenneke
,
K.
Schwarzkopf
,
A.
Manz
, and
M.
Schneider
, “
Microfluidics as tool to prepare size-tunable PLGA nanoparticles with high curcumin encapsulation for efficient mucus penetration
,”
Beilstein J. Nanotechnol.
10
,
2280
2293
(
2019
) (in English).
50.
R.
Donno
et al, “
Nanomanufacturing through microfluidic-assisted nanoprecipitation: Advanced analytics and structure-activity relationships
,”
Int. J. Pharm.
534
(
1
),
97
107
(
2017
).
51.
S.
Rezvantalab
and
M. K.
Moraveji
, “
Microfluidic assisted synthesis of PLGA drug delivery systems
,”
RSC Adv.
9
(
4
),
2055
2072
(
2019
).
52.
S.
Gimondi
et al, “
Microfluidic mixing system for precise PLGA-PEG nanoparticles size control
,”
Nanomed.: Nanotechnol. Biol. Med.
40
,
102482
(
2022
).
53.
F.
Danhier
,
E.
Ansorena
,
J. M.
Silva
,
R.
Coco
,
A. L.
Breton
, and
V.
Préat
, “
PLGA-based nanoparticles: An overview of biomedical applications
,”
J. Controlled Release
161
(
2
),
505
522
(
2012
).
54.
E.
Sah
and
H.
Sah
, “
Recent trends in preparation of poly(lactide-co-glycolide) nanoparticles by mixing polymeric organic solution with antisolvent
,”
J. Nanomater.
2015
,
794601
(
2015
).
55.
l.-J.
Pan
et al, “
Controllable synthesis of nanocrystals in droplet reactors
,”
Lab Chip
18
(
1
),
41
56
(
2018
).
56.
H.-H.
Jeong
,
V. R.
Yelleswarapu
,
S.
Yadavali
,
D.
Issadore
, and
D.
Lee
, “
Kilo-scale droplet generation in three-dimensional monolithic elastomer device (3D MED)
,”
Lab Chip
15
(
23
),
4387
4392
(
2015
).
57.
A. M.
Nightingale
et al, “
A stable droplet reactor for high temperature nanocrystal synthesis
,”
Lab Chip
11
(
7
),
1221
1227
(
2011
).
58.
P.
Zhao
et al, “
Long-term storage of lipid-like nanoparticles for mRNA delivery
,”
Bioactive Mater.
5
(
2
),
358
363
(
2020
).
59.
J.
Kim
,
Y.
Eygeris
,
M.
Gupta
, and
G.
Sahay
, “
Self-assembled mRNA vaccines
,”
Adv. Drug Delivery Rev.
170
,
83
112
(
2021
).
60.
P.
Arosio
,
T.
Müller
,
L.
Mahadevan
, and
T. P. J.
Knowles
, “
Density-gradient-free microfluidic centrifugation for analytical and preparative separation of nanoparticles
,”
Nano Lett.
14
(
5
),
2365
2371
(
2014
).
61.
M. A.
Livshits
et al, “
Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol
,”
Sci. Rep.
5
(
1
),
17319
(
2015
).
62.
J.
Lohwacharin
and
S.
Takizawa
, “
Effects of nanoparticles on the ultrafiltration of surface water
,”
J. Membr. Sci.
326
(
2
),
354
362
(
2009
).
63.
G.-T.
Wei
and
F.-K.
Liu
, “
Separation of nanometer gold particles by size exclusion chromatography
,”
J. Chromatogr., A
836
(
2
),
253
260
(
1999
).
64.
K. Y.
Chung
,
J. M.
Quek
,
S. H.
Neo
, and
H. P.
Too
, “
Polymer-based precipitation of extracellular vesicular miRNAs from serum improve gastric cancer miRNA biomarker performance
,”
J. Mol. Diagn.
22
(
5
),
610
618
(
2020
).
65.
D.
Brambilla
et al, “
EV separation: Release of intact extracellular vesicles immunocaptured on magnetic particles
,”
Anal. Chem.
93
(
13
),
5476
5483
(
2021
) (in English).
66.
S.
Zhao
et al, “
A disposable acoustofluidic chip for nano/microparticle separation using unidirectional acoustic transducers
,”
Lab Chip
20
(
7
),
1298
1308
(
2020
).
67.
M.
Dimaki
,
M. H.
Olsen
,
N.
Rozlosnik
, and
W. E.
Svendsen
, “
Sub-100 nm nanoparticle upconcentration in flow by dielectrophoretic forces
,”
Micromachines
13
(
6
),
866
(
2022
) (in English).
68.
S.
Khizar
,
H.
Ben Halima
,
N. M.
Ahmad
,
N.
Zine
,
A.
Errachid
, and
A.
Elaissari
, “
Magnetic nanoparticles in microfluidic and sensing: From transport to detection
,”
Electrophoresis
41
(
13–14
),
1206
1224
(
2020
) (in English).
69.
H.
Zhao
et al, “
Continuous optical sorting of nanoscale biomolecules in integrated microfluidic-nanophotonic chips
,”
Sens. Actuators, B
331
,
129428
(
2021
).
70.
Q.
Zhao
,
D.
Yuan
,
J.
Zhang
, and
W.
Li
, “
A review of secondary flow in inertial microfluidics
,”
Micromachines
11
(
5
),
461
(
2020
).
71.
M.
Al-Fandi
,
M.
Al-Rousan
,
M. A. K.
Jaradat
, and
L.
Al-Ebbini
, “
New design for the separation of microorganisms using microfluidic deterministic lateral displacement
,”
Rob. Comput. Integ. Manuf.
27
(
2
),
237
244
(
2011
).
72.
Y.
Cheng
,
Y.
Wang
,
Z.
Ma
,
W.
Wang
, and
X.
Ye
, “
A bubble- and clogging-free microfluidic particle separation platform with multi-filtration
,”
Lab Chip
16
(
23
),
4517
4526
(
2016
).
73.
A. L.
Vig
and
A.
Kristensen
, “
Separation enhancement in pinched flow fractionation
,”
Appl. Phys. Lett.
93
(
20
),
203507
(
2008
).
74.
G.
Romeo
,
G.
D’Avino
,
F.
Greco
,
P. A.
Netti
, and
P. L.
Maffettone
, “
Viscoelastic flow-focusing in microchannels: Scaling properties of the particle radial distributions
,”
Lab Chip
13
(
14
),
2802
2807
(
2013
).
75.
S.
Yan
et al, “
An integrated dielectrophoresis-active hydrophoretic microchip for continuous particle filtration and separation
,”
J. Micromech. Microeng.
25
(
8
),
084010
(
2015
).
76.
A. M.
Barbuti
and
Z. S.
Chen
, “
Paclitaxel through the ages of anticancer therapy: Exploring its role in chemoresistance and radiation therapy
,”
Cancers
7
(
4
),
2360
2371
(
2015
) (in English).
77.
Y.
Oduk
et al, “
VEGF nanoparticles repair the heart after myocardial infarction
,”
Am. J. Physiol. Heart Circ. Physiol.
314
(
2
),
H278
H284
(
2017
).
78.
X.
Hou
,
T.
Zaks
,
R.
Langer
, and
Y.
Dong
, “
Lipid nanoparticles for mRNA delivery
,”
Nat. Rev. Mater.
6
(
12
),
1078
1094
(
2021
) (in English).
79.
M. M.
El-Hammadi
and
J. L.
Arias
, “
Recent advances in the surface functionalization of PLGA-based nanomedicines
,”
Nanomaterials
12
(
3
),
354
(
2022
) (in English).
80.
l.
Xin
,
H.-T.
Zhang
,
W.-F.
Yang
,
Y.-F.
Li
, and
C.
Liu
, “
Evaluation of METase-pemetrexed-loaded PEG–PLGA nanoparticles modified with anti-CD133–scFV for treatment of gastric carcinoma
,”
Biosci. Rep.
38
(
1
),
BSR20171001
(
2018
).
81.
W.
Hariri
,
T.
Sudha
,
D. J.
Bharali
,
H.
Cui
, and
S. A.
Mousa
, “
Nano-targeted delivery of toremifene, an estrogen receptor-α blocker in prostate cancer
,”
Pharm. Res.
32
,
2764
2774
(
2015
).
82.
R.
Domínguez-Ríos
et al, “
Cisplatin-loaded PLGA nanoparticles for HER2 targeted ovarian cancer therapy
,”
Colloids Surf., B
178
,
199
207
(
2019
).
83.
X.
Zhang
et al, “
Trastuzumab-coated nanoparticles loaded with docetaxel for breast cancer therapy
,”
Dose-Response
17
(
3
),
1559325819872583
(
2019
).
84.
Z.
Zhou
,
A.
Badkas
,
M.
Stevenson
,
J.-Y.
Lee
, and
Y.-K.
Leung
, “
Herceptin conjugated PLGA-PHis-PEG pH sensitive nanoparticles for targeted and controlled drug delivery
,”
Int. J. Pharm.
487
(
1–2
),
81
90
(
2015
).
85.
M.
Mehdizadeh
et al, “
Biotin decorated PLGA nanoparticles containing SN-38 designed for cancer therapy
,”
Artif. Cells, Nanomed., Biotechnol.
45
(
3
),
495
504
(
2017
).
86.
E.
Cenni
et al, “
The effect of poly (d, l-lactide-co-glycolide)-alendronate conjugate nanoparticles on human osteoclast precursors
,”
J. Biomater. Sci., Polym. Ed.
23
(
10
),
1285
1300
(
2012
).
87.
K. R.
Chaudhari
et al, “
Bone metastasis targeting: A novel approach to reach bone using zoledronate anchored PLGA nanoparticle as carrier system loaded with Docetaxel
,”
J. Controlled Release
158
(
3
),
470
478
(
2012
).
88.
J.
Zhang
et al, “
Development of drug loaded nanoparticles binding to hydroxyapatite based on a bisphosphonate modified nonionic surfactant
,”
J. Nanomater.
2015
(
1
),
393968
(
2015
).
89.
R.
Pieroth
,
S.
Paver
,
S.
Day
, and
C.
Lammersfeld
, “
Folate and its impact on cancer risk
,”
Curr. Nutr. Rep.
7
,
70
84
(
2018
).
90.
M. L.
Bruschi
, “
Lectins and nanostructured drug delivery systems
,”
Curr. Drug Delivery
16
(
3
),
268
269
(
2019
).
91.
J.
Chen
et al, “
Solanum tuberosum lectin-conjugated PLGA nanoparticles for nose-to-brain delivery: In vivo and in vitro evaluations
,”
J. Drug Targeting
20
(
2
),
174
184
(
2012
).
92.
S. K.
Jain
,
T.
Haider
,
A.
Kumar
, and
A.
Jain
, “
Lectin-conjugated clarithromycin and acetohydroxamic acid-loaded PLGA nanoparticles: A novel approach for effective treatment of H. pylori
,”
AAPS PharmSciTech
17
,
1131
1140
(
2016
).
93.
Z.
Wen
et al, “
Odorranalectin-conjugated nanoparticles: Preparation, brain delivery and pharmacodynamic study on Parkinson's disease following intranasal administration
,”
J. Controlled Release
151
(
2
),
131
138
(
2011
).
94.
G.
Wu
,
F.
Zhou
,
L.
Ge
,
X.
Liu
, and
F.
Kong
, “
Novel mannan-PEG-PE modified bioadhesive PLGA nanoparticles for targeted gene delivery
,”
J. Nanomater.
,
2012
,
1
, p.
981670
,
2012
.
95.
J.
Kaur
and
K.
Tikoo
, “
Ets1 identified as a novel molecular target of RNA aptamer selected against metastatic cells for targeted delivery of nano-formulation
,”
Oncogene
34
(
41
),
5216
5228
(
2015
).
96.
I.
Monaco
et al, “
Aptamer functionalization of nanosystems for glioblastoma targeting through the blood–brain barrier
,”
J. Med. Chem.
60
(
10
),
4510
4516
(
2017
).
97.
M.
Ni
et al, “
Poly (lactic-co-glycolic acid) nanoparticles conjugated with CD133 aptamers for targeted salinomycin delivery to CD133+ osteosarcoma cancer stem cells
,”
Int. J. Nanomed.
10
(
1
),
2537
2554
(
2015
).
98.
G.
Ravichandran
and
A. K.
Rengan
, “
Aptamer-mediated nanotheranostics for cancer treatment: A review
,”
ACS Appl. Nano Mater.
3
(
10
),
9542
9559
(
2020
).
99.
M.
Alipour
et al, “
Recent progress in biomedical applications of RGD-based ligand: From precise cancer theranostics to biomaterial engineering: A systematic review
,”
J. Biomed. Mater. Res. A
108
(
4
),
839
850
(
2020
).
100.
Y.
Cui
et al, “
Dual-target peptide-modified erythrocyte membrane-enveloped PLGA nanoparticles for the treatment of glioma
,”
Front. Oncol.
10
,
563938
(
2020
).
101.
N.
Feiner-Gracia
,
A.
Dols-Pérez
,
M.
Royo
,
C.
Solans
,
M.
Garcia-Celma
, and
C.
Fornaguera
, “
Cell penetrating peptide grafting of PLGA nanoparticles to enhance cell uptake
,”
Eur. Polym. J.
108
,
429
438
(
2018
).
102.
I.
Gessner
and
I.
Neundorf
, “
Nanoparticles modified with cell-penetrating peptides: Conjugation mechanisms, physicochemical properties, and application in cancer diagnosis and therapy
,”
Int. J. Mol. Sci.
21
(
7
),
2536
(
2020
).
103.
R.
Gonzalez-Pizarro
et al, “
Ocular penetration of fluorometholone-loaded PEG-PLGA nanoparticles functionalized with cell-penetrating peptides
,”
Nanomedicine
14
(
23
),
3089
3104
(
2019
).
104.
G. P.
Hoyos-Ceballos
et al, “
PLGA-PEG-ANG-2 nanoparticles for blood–brain barrier crossing: Proof-of-concept study
,”
Pharmaceutics
12
(
1
),
72
(
2020
).
105.
R. L.
Manthe
and
S.
Muro
, “
ICAM-1-targeted nanocarriers attenuate endothelial release of soluble ICAM-1, an inflammatory regulator
,”
Bioeng. Transl. Med.
2
(
1
),
109
119
(
2017
).
106.
N.
Mehrotra
,
S.
Kharbanda
, and
H.
Singh
, “
Peptide-based combination nanoformulations for cancer therapy
,”
Nanomedicine
15
(
22
),
2201
2217
(
2020
).
107.
X.
Nie
et al, “
SP94 peptide-functionalized PEG-PLGA nanoparticle loading with cryptotanshinone for targeting therapy of hepatocellular carcinoma
,”
AAPS PharmSciTech
21
,
1
11
(
2020
).
108.
J. M.
Rios De
L.
Rosa
et al, “
Microfluidic-assisted preparation of RGD-decorated nanoparticles: Exploring integrin-facilitated uptake in cancer cell lines
,”
Sci. Rep.
10
(
1
),
14505
(
2020
).
109.
C.
Chittasupho
,
S.-X.
Xie
,
A.
Baoum
,
T.
Yakovleva
,
T. J.
Siahaan
, and
C. J.
Berkland
, “
ICAM-1 targeting of doxorubicin-loaded PLGA nanoparticles to lung epithelial cells
,”
Eur. J. Pharm. Sci.
37
(
2
),
141
150
(
2009
).
110.
H.
Choudhury
et al, “
Transferrin receptors-targeting nanocarriers for efficient targeted delivery and transcytosis of drugs into the brain tumors: A review of recent advancements and emerging trends
,”
Drug Delivery Transl. Res.
8
,
1545
1563
(
2018
).
111.
I.
Aranaz
et al, “
Chitosan: An overview of Its properties and applications
,”
Polymers
13
(
19
),
3256
(
2021
).
112.
R. J. C.
Bose
,
S.-H.
Lee
, and
H.
Park
, “
Lipid-based surface engineering of PLGA nanoparticles for drug and gene delivery applications
,”
Biomater. Res.
20
(
1
),
34
(
2016
).
113.
B.
Mandal
et al.
, “
Core–shell-type lipid–polymer hybrid nanoparticles as a drug delivery platform
,”
Nanomed. Nanotechnol. Biol. Med.
9
(
4
),
474
491
(
2013
).
114.
Q.
Zhong
et al, “
Optimization of DNA delivery by three classes of hybrid nanoparticle/DNA complexes
,”
J. Nanobiotechnol.
8
(
1
),
6
(
2010
).
115.
K.
Hadinoto
,
A.
Sundaresan
, and
W. S.
Cheow
, “
Lipid–polymer hybrid nanoparticles as a new generation therapeutic delivery platform: A review
,”
Eur. J. Pharm. Biopharm.
85
(
3
),
427
443
(
2013
).
116.
C.
Clawson
,
L.
Ton
,
S.
Aryal
,
V.
Fu
,
S.
Esener
, and
L.
Zhang
, “
Synthesis and characterization of lipid–polymer hybrid nanoparticles with pH-triggered poly(ethylene glycol) shedding
,”
Langmuir
27
(
17
),
10556
10561
(
2011
).
117.
C.
Hald Albertsen
,
J. A.
Kulkarni
,
D.
Witzigmann
,
M.
Lind
,
K.
Petersson
, and
J. B.
Simonsen
, “
The role of lipid components in lipid nanoparticles for vaccines and gene therapy
,”
Adv. Drug Delivery Rev.
188
,
114416
(
2022
).
118.
E.
Fröhlich
, “
The role of surface charge in cellular uptake and cytotoxicity of medical nanoparticles
,”
Int. J. Nanomed.
7
,
5577
5591
(
2012
) (in English).
119.
l.
Shi
et al, “
Effects of polyethylene glycol on the surface of nanoparticles for targeted drug delivery
,”
Nanoscale
13
(
24
),
10748
10764
(
2021
).
120.
T.
Miyazawa
,
M.
Itaya
,
G. C.
Burdeos
,
K.
Nakagawa
, and
T.
Miyazawa
, “
A critical review of the use of surfactant-coated nanoparticles in nanomedicine and food nanotechnology
,”
Int. J. Nanomed.
16
,
3937
3999
(
2021
) (in English).
121.
A.
Pucci
,
E.
Locatelli
,
J.
Ponti
,
C.
Uboldi
,
V.
Molinari
, and
M.
Comes Franchini
, “
Click chemistry on the surface of PLGA-b-PEG polymeric nanoparticles: A novel targetable fluorescent imaging nanocarrier
,”
J. Nanopart. Res.
15
,
1
6
(
2013
).
122.
M.
Esfandyari-Manesh
,
M.
Abdi
,
A. H.
Talasaz
,
S. M.
Ebrahimi
,
F.
Atyabi
, and
R.
Dinarvand
, “
S2P peptide-conjugated PLGA-Maleimide-PEG nanoparticles containing Imatinib for targeting drug delivery to atherosclerotic plaques
,”
Daru, J. Pharm. Sci.
28
,
131
138
(
2020
).
123.
I.
Slowing
,
B. G.
Trewyn
, and
V. S. Y.
Lin
, “
Effect of surface functionalization of MCM-41-type mesoporous silica nanoparticles on the endocytosis by human cancer cells
,”
J. Am. Chem. Soc.
128
(
46
),
14792
14793
(
2006
).
124.
D. L. J.
Thorek
and
A.
Tsourkas
, “
Size, charge and concentration dependent uptake of iron oxide particles by non-phagocytic cells
,”
Biomaterials
29
(
26
),
3583
3590
(
2008
).
125.
J. V.
Jokerst
,
T.
Lobovkina
,
R. N.
Zare
, and
S. S.
Gambhir
, “
Nanoparticle PEGylation for imaging and therapy
,”
Nanomedicine
6
(
4
),
715
728
(
2011
).
126.
H.
Lee
,
H.
Fonge
,
B.
Hoang
,
R. M.
Reilly
, and
C.
Allen
, “
The effects of particle size and molecular targeting on the intratumoral and subcellular distribution of polymeric nanoparticles
,”
Mol. Pharm.
7
(
4
),
1195
1208
(
2010
).
127.
H.
Shmeeda
,
D.
Tzemach
,
L.
Mak
, and
A.
Gabizon
, “
Her2-targeted pegylated liposomal doxorubicin: Retention of target-specific binding and cytotoxicity after in vivo passage
,”
J. Controlled Release
136
(
2
),
155
160
(
2009
).
128.
M.
Wang
and
M.
Thanou
, “
Targeting nanoparticles to cancer
,”
Pharmacol. Res.
62
(
2
),
90
99
(
2010
).
129.
M. J.
Mitchell
,
M. M.
Billingsley
,
R. M.
Haley
,
M. E.
Wechsler
,
N. A.
Peppas
, and
R.
Langer
, “
Engineering precision nanoparticles for drug delivery
,”
Nat. Rev. Drug Discovery
20
(
2
),
101
124
(
2021
).
130.
N.
Hoshyar
,
S.
Gray
,
H.
Han
, and
G.
Bao
, “
The effect of nanoparticle size on in vivo pharmacokinetics and cellular interaction
,”
Nanomedicine
11
(
6
),
673
692
(
2016
).
131.
G.
Adamo
,
S.
Campora
, and
G.
Ghersi
, “
Functionalization of nanoparticles in specific targeting and mechanism release
,” in
Nanostructures for Novel Therapy
, edited by
D.
Ficai
and
A. M.
Grumezescu
(
Elsevier
,
2017
), Chap. III, pp.
57
80
.
132.
V.
Schäfer
et al, “
Phagocytosis of nanoparticles by human immunodeficiency virus (HlV)-infected macrophages: A possibility for antiviral drug targeting
,”
Pharm. Res.
9
(
4
),
541
546
(
1992
).
133.
H.
Cabral
et al, “
Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours depends on size
,”
Nat. Nanotechnol.
6
(
12
),
815
823
(
2011
).
134.
W. M.
MacCuaig
et al, “
Active targeting significantly outperforms nanoparticle size in facilitating tumor-specific uptake in orthotopic pancreatic cancer
,”
ACS Appl. Mater. Interfaces
13
(
42
),
49614
49630
(
2021
) (in English).
135.
W.
Jiang
,
B. Y. S.
Kim
,
J. T.
Rutka
, and
W. C. W.
Chan
, “
Nanoparticle-mediated cellular response is size-dependent
,”
Nat. Nanotechnol.
3
(
3
),
145
150
(
2008
).
136.
E.
Blanco
,
H.
Shen
, and
M.
Ferrari
, “
Principles of nanoparticle design for overcoming biological barriers to drug delivery
,”
Nat. Biotechnol.
33
(
9
),
941
951
(
2015
).
137.
A.
Aderem
and
D. M.
Underhill
, “
Mechanisms of phagocytosis in macrophages
,”
Annu. Rev. Immunol.
17
(
1
),
593
623
(
1999
).
138.
l. A.
Carver
and
J. E.
Schnitzer
, “
Caveolae: Mining little caves for new cancer targets
,”
Nat. Rev. Cancer
3
(
8
),
571
581
(
2003
).
139.
G. J.
Doherty
and
H. T.
McMahon
, “
Mechanisms of endocytosis
,”
Annu. Rev. Biochem.
78
(
1
),
857
902
(
2009
).
140.
M.
Ehrlich
et al, “
Endocytosis by random initiation and stabilization of clathrin-coated pits
,”
Cell
118
(
5
),
591
605
(
2004
).
141.
l. E.
Kelemen
, “
The role of folate receptor α in cancer development, progression and treatment: Cause, consequence or innocent bystander?
,”
Int. J. Cancer
119
(
2
),
243
250
(
2006
).
142.
Y.
Lu
and
P. S.
Low
, “
Folate-mediated delivery of macromolecular anticancer therapeutic agents
,”
Adv. Drug Delivery Rev.
64
,
342
352
(
2012
).
143.
J. P.
Lim
and
P. A.
Gleeson
, “
Macropinocytosis: An endocytic pathway for internalising large gulps
,”
Immunol. Cell Biol.
89
(
8
),
836
843
(
2011
).
144.
S. E. A.
Gratton
et al, “
The effect of particle design on cellular internalization pathways
,”
Proc. Natl. Acad. Sci. U.S.A.
105
(
33
),
11613
11618
(
2008
).
145.
B. D.
Chithrani
,
A. A.
Ghazani
, and
W. C. W.
Chan
, “
Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells
,”
Nano Lett.
6
(
4
),
662
668
(
2006
).
146.
Y.
Qiu
et al, “
Surface chemistry and aspect ratio mediated cellular uptake of Au nanorods
,”
Biomaterials
31
(
30
),
7606
7619
(
2010
).
147.
B.
Zhang
,
P.
Sai Lung
,
S.
Zhao
,
Z.
Chu
,
W.
Chrzanowski
, and
Q.
Li
, “
Shape dependent cytotoxicity of PLGA-PEG nanoparticles on human cells
,”
Sci. Rep.
7
(
1
),
7315
(
2017
).
148.
C. Y.
Hsu
,
P. W.
Wang
,
A.
Alalaiwe
,
Z. C.
Lin
, and
J. Y.
Fang
, “
Use of lipid nanocarriers to improve oral delivery of vitamins
,”
Nutrients
11
(
1
),
68
(
2019
) (in English).
149.
H.
Jin
,
D. A.
Heller
,
R.
Sharma
, and
M. S.
Strano
, “
Size-dependent cellular uptake and expulsion of single-walled carbon nanotubes: Single particle tracking and a generic uptake model for nanoparticles
,”
ACS Nano
3
(
1
),
149
158
(
2009
).
150.
J.
Wang
,
S.
Tian
,
R. A.
Petros
,
M. E.
Napier
, and
J. M.
DeSimone
, “
The complex role of multivalency in nanoparticles targeting the transferrin receptor for cancer therapies
,”
J. Am. Chem. Soc.
132
(
32
),
11306
11313
(
2010
).
151.
C. J.
Martínez Rivas
et al, “
Nanoprecipitation process: From encapsulation to drug delivery
,”
Int. J. Pharm.
532
(
1
),
66
81
(
2017
).
152.
S.
Kumar
,
N.
Dilbaghi
,
R.
Rani
, and
G.
Bhanjana
, “
Nanotechnology as emerging tool for enhancing solubility of poorly water-soluble drugs
,”
BioNanoScience
2
,
227
(
2012
).
153.
M. T.
Peracchia
,
R.
Gref
,
Y.
Minamitake
,
A.
Domb
,
N.
Lotan
, and
R.
Langer
, “
PEG-coated nanospheres from amphiphilic diblock and multiblock copolymers: Investigation of their drug encapsulation and release characteristics
,”
J. Controlled Release
46
(
3
),
223
231
(
1997
).
154.
G.
Yurtdaş Kırımlıoğlu
, “
Drug loading methods and drug release mechanisms of PLGA nanoparticles
,” in
Poly(Lactic-co-Glycolic Acid) (PLGA) Nanoparticles for Drug Delivery
, edited by
P.
Kesharwani
(
Elsevier
,
2023
), Chap. III, pp.
55
86
.
155.
H.
Safari
,
M. L.
Felder
,
N.
Kaczorowski
, and
O.
Eniola-Adefeso
, “
Effect of the emulsion solvent evaporation technique cosolvent choice on the loading efficiency and release profile of anti-CD47 from PLGA nanospheres
,”
J. Pharm. Sci.
111
(
9
),
2525
2530
(
2022
).
156.
M.
Jelvehgari
and
S. H.
Montazam
, “
Comparison of microencapsulation by emulsion-solvent extraction/evaporation technique using derivatives cellulose and acrylate-methacrylate copolymer as carriers
,”
Jundishapur J. Nat. Pharm. Prod.
7
(
4
),
144
152
(
2012
) (in English).
157.
Y.
Xu
,
C. S.
Kim
,
D. M.
Saylor
, and
D.
Koo
, “
Polymer degradation and drug delivery in PLGA-based drug–polymer applications: A review of experiments and theories
,”
J. Biomed. Mater. Res. B
105
(
6
),
1692
1716
(
2017
).
158.
S. M.
Bennett
et al, “
The effect of particle size on the in vivo degradation of poly (d, l-lactide-co-glycolide)/α-tricalcium phosphate micro- and nanocomposites
,”
Acta Biomater.
45
,
340
348
(
2016
).
159.
T.
Bourguignon
,
A. A.
Torrano
,
L.
Houel-Renault
,
A.
Machelart
,
P.
Brodin
, and
R.
Gref
, “
An original methodology to study polymeric nanoparticle-macrophage interactions: Nanoparticle tracking analysis in cell culture media and quantification of the internalized objects
,”
Int. J. Pharm.
610
,
121202
(
2021
).
160.
F.
Esmaeili
,
M. H.
Ghahremani
,
B.
Esmaeili
,
M. R.
Khoshayand
,
F.
Atyabi
, and
R.
Dinarvand
, “
PLGA nanoparticles of different surface properties: Preparation and evaluation of their body distribution
,”
Int. J. Pharm.
349
(
1–2
),
249
255
(
2008
).
161.
J.-W.
Yoo
,
E.
Chambers
, and
S.
Mitragotri
, “
Factors that control the circulation time of nanoparticles in blood: Challenges, solutions and future prospects
,”
Curr. Pharm. Des.
16
(
21
),
2298
2307
(
2010
).
162.
M.
Arnida
,
A.
Ray
,
C.
Peterson
, and
H.
Ghandehari
, “
Geometry and surface characteristics of gold nanoparticles influence their biodistribution and uptake by macrophages
,”
Eur. J. Pharm. Biopharm.
77
(
3
),
417
423
(
2011
).
163.
A.
Albanese
,
P. S.
Tang
, and
W. C.
Chan
, “
The effect of nanoparticle size, shape, and surface chemistry on biological systems
,”
Annu. Rev. Biomed. Eng.
14
,
1
16
(
2012
).
164.
K.
Knop
,
R.
Hoogenboom
,
D.
Fischer
, and
U. S.
Schubert
, “
Poly(ethylene glycol) in drug delivery: Pros and cons as well as potential alternatives
,”
Angew. Chem., Int. Ed.
49
(
36
),
6288
6308
(
2010
).
165.
A. S.
Abu Lila
,
H.
Kiwada
, and
T.
Ishida
, “
The accelerated blood clearance (ABC) phenomenon: Clinical challenge and approaches to manage
,”
J. Controlled Release
172
(
1
),
38
47
(
2013
).
166.
M. A.
Maier
et al, “
Biodegradable lipids enabling rapidly eliminated lipid nanoparticles for systemic delivery of RNAi therapeutics
,”
Mol. Ther.
21
(
8
),
1570
1578
(
2013
).
167.
S.
Sabnis
et al, “
A novel amino lipid series for mRNA delivery: Improved endosomal escape and sustained pharmacology and safety in non-human primates
,”
Mol. Ther.
26
(
6
),
1509
1519
(
2018
).
168.
K. J.
Hassett
et al, “
Optimization of lipid nanoparticles for intramuscular administration of mRNA vaccines
,”
Mol. Ther. Nucl. Acids
15
,
1
11
(
2019
).
169.
X.
Zhang
et al, “
Functionalized lipid-like nanoparticles for in vivo mRNA delivery and base editing
,”
Sci. Adv.
6
(
34
),
eabc2315
(
2020
).
170.
X.
Zhang
et al, “
Biodegradable amino-ester nanomaterials for Cas9 mRNA delivery in vitro and in vivo
,”
ACS Appl. Mater. Interfaces
9
(
30
),
25481
25487
(
2017
).
171.
J. S.
Suk
,
Q.
Xu
,
N.
Kim
,
J.
Hanes
, and
L. M.
Ensign
, “
PEGylation as a strategy for improving nanoparticle-based drug and gene delivery
,”
Adv. Drug Delivery Rev.
99
,
28
51
(
2016
).
172.
J.
Kim
,
H. J.
Vaughan
,
C. G.
Zamboni
,
J. C.
Sunshine
, and
J. J.
Green
, “
High-throughput evaluation of polymeric nanoparticles for tissue-targeted gene expression using barcoded plasmid DNA
,”
J. Controlled Release
337
,
105
116
(
2021
).
173.
S.
Mura
et al, “
Influence of surface charge on the potential toxicity of PLGA nanoparticles towards Calu-3 cells
,”
Int. J. Nanomed.
6
(
null
),
2591
2605
(
2011
).
174.
G.
Martínez
et al, “
Environmental impact of nanoparticles’ application as an emerging technology: A review
,”
Materials
14
(
1
),
166
(
2020
) (in English).
175.
S.
Rajkovic
,
N. A.
Bornhöft
,
R.
van der Weijden
,
B.
Nowack
, and
V.
Adam
, “
Dynamic probabilistic material flow analysis of engineered nanomaterials in European waste treatment systems
,”
Waste Manage.
113
,
118
131
(
2020
).
176.
B.
Nowack
et al, “
Potential scenarios for nanomaterial release and subsequent alteration in the environment
,”
Environ. Toxicol. Chem.
31
(
1
),
50
59
(
2012
).
177.
A. L.
Dale
,
E. A.
Casman
,
G. V.
Lowry
,
J. R.
Lead
,
E.
Viparelli
, and
M.
Baalousha
, “
Modeling nanomaterial environmental fate in aquatic systems
,”
Environ. Sci. Technol.
49
(
5
),
2587
2593
(
2015
).
178.
B.
Nowack
, “
Evaluation of environmental exposure models for engineered nanomaterials in a regulatory context
,”
NanoImpact
8
,
38
47
(
2017
).
179.
A.
Elsaesser
and
C. V.
Howard
, “
Toxicology of nanoparticles
,”
Adv. Drug Delivery Rev.
64
(
2
),
129
137
(
2012
).
180.
J.
Lojk
,
J.
Repas
,
P.
Veranič
,
V. B.
Bregar
, and
M.
Pavlin
, “
Toxicity mechanisms of selected engineered nanoparticles on human neural cells in vitro
,”
Toxicology
432
,
152364
(
2020
).
181.
M. J.
Akhtar
,
M.
Ahamed
,
H.
Alhadlaq
, and
S.
Alrokayan
, “
Toxicity mechanism of gadolinium oxide nanoparticles and gadolinium ions in human breast cancer cells
,”
Curr. Drug Metab.
20
(
11
),
907
917
(
2019
).
182.
M.
Ghosh
,
M.
Bandyopadhyay
, and
A.
Mukherjee
, “
Genotoxicity of titanium dioxide (TiO2) nanoparticles at two trophic levels: Plant and human lymphocytes
,”
Chemosphere
81
(
10
),
1253
1262
(
2010
).
183.
R.
Lehner
,
C.
Weder
,
A.
Petri-Fink
, and
B.
Rothen-Rutishauser
, “
Emergence of nanoplastic in the environment and possible impact on human health
,”
Environ. Sci. Technol.
53
(
4
),
1748
1765
(
2019
).
184.
J.
Wang
et al, “
Disruption of zebrafish (Danio rerio) reproduction upon chronic exposure to TiO2 nanoparticles
,”
Chemosphere
83
(
4
),
461
467
(
2011
).
185.
K. P.
Steckiewicz
et al, “
Impact of gold nanoparticles shape on their cytotoxicity against human osteoblast and osteosarcoma in in vitro model. Evaluation of the safety of use and anti-cancer potential
,”
J. Mater. Sci.: Mater. Med.
30
(
2
),
22
(
2019
).
186.
C. J.
Labrador-Rached
,
R. T.
Browning
,
L. K.
Braydich-Stolle
, and
K. K.
Comfort
, “
Toxicological implications of platinum nanoparticle exposure: Stimulation of intracellular stress, inflammatory response, and Akt signaling in vitro
,”
J. Toxicol.
2018
,
1367801
(
2018
).