Next Article in Journal
Bacterial Cellulose-Derived Sorbents for Cr (VI) Remediation: Adsorption, Elution, and Reuse
Previous Article in Journal
A Review on the Application of Deep Eutectic Solvents in Polymer-Based Membrane Preparation for Environmental Separation Technologies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Applications of PLGA in Drug Delivery Systems

1
Department of Burns & Plastic Surgery, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou 510006, China
2
College of Pharmacy, Jinan University, Guangzhou 510006, China
3
International School, Jinan University, Guangzhou 510006, China
4
Guangdong Institute for Drug Control, NMPA Key Laboratory for Quality Control and Evaluation of Pharmaceutical Excipients, Guangzhou 510660, China
*
Author to whom correspondence should be addressed.
Polymers 2024, 16(18), 2606; https://doi.org/10.3390/polym16182606
Submission received: 21 June 2024 / Revised: 18 August 2024 / Accepted: 11 September 2024 / Published: 14 September 2024

Abstract

:
Poly(lactic-co-glycolic acid) (PLGA) is a widely used biodegradable and biocompatible copolymer in drug delivery systems (DDSs). In this article, we highlight the critical physicochemical properties of PLGA, including its molecular weight, intrinsic viscosity, monomer ratio, blockiness, and end caps, that significantly influence drug release profiles and degradation times. This review also covers the extensive literature on the application of PLGA in delivering small-molecule drugs, proteins, peptides, antibiotics, and antiviral drugs. Furthermore, we discuss the role of PLGA-based DDSs in the treating various diseases, including cancer, neurological disorders, pain, and inflammation. The incorporation of drugs into PLGA nanoparticles and microspheres has been shown to enhance their therapeutic efficacy, reduce toxicity, and improve patient compliance. Overall, PLGA-based DDSs holds great promise for the advancement of the treatment and management of multiple chronic conditions.

Graphical Abstract

1. Introduction

Drug delivery systems (DDSs) are designed to deliver drugs to specific target sites, offering many advantages, including enhanced therapeutic efficacy, improved patient compliance, reduced dose-related toxicity, and support for a broader range of medical treatments [1]. The routes of DDSs vary depending on the specific treatment, whether oral, pulmonary, injectable, or transdermal, and implantation method [2].
Controlled drug delivery (CDD), which can be categorized into passive and active-controlled DDSs, releases drugs at a predetermined rate and for a predetermined duration to maintain therapeutic drug levels in the body while improving efficacy and minimizing side effects [3,4]. Compared to traditional DDSs, CDD systems are particularly beneficial for drugs with narrow therapeutic windows that require frequent monitoring and adjustment or for long-term treatments [5]. Long-acting drug delivery systems (LADDSs) continuously release active pharmaceutical ingredients (APIs) over periods ranging from weeks to years, revolutionizing the management and treatment of multiple chronic conditions, including cancer [6], neurological disorders [7], pain [8], inflammation [9], and other diseases [10]. LADDSs also offer the benefits of reducing the frequency of injections, minimizing administration pain [11], and improving patient comfort [12].
Various materials are used for formulations to achieve long-acting delivery and controlled drug release, such as homopolymers of poly(lactide) or poly(lactic acid) (PLA), polyester copolymers of lactide and glycolide (PLG), and copolymers of lactic acid and glycolic acid (PLGA) [13]. The molecular weight and monomer ratio of these polyester copolymers play a crucial role in determining the duration of sustained release time. PLGA, which is widely used due to its excellent biocompatibility and biodegradability, is commonly utilized in DDSs. PLGA-based DDSs can be tailored to release drugs at controlled rates, protect them from degradation, and enhance stability throughout the product life cycle [14]. PLGA can be synthesized through direct condensation of lactic acid and glycolic acid or by ring-opening polymerization (ROP) of lactide and glycolide cyclic dimers. Initiators like tin (II) bis (2-ethyl hexanoate) (Sn (Oct)2), zinc proline, and stannous octoate are used in ROP to obtain PLGA copolymers of the desired weight with hydroxyl and carboxyl end groups [15,16,17]. Meanwhile, PLGA can be modified with functional polymeric blocks such as polyethylene glycol (PEG) [18].
In an aqueous environment, PLGA degradation occurs due to multiple factors, including the hydrolysis of ester linkages, decreased molecular weight, and heterogeneous erosion [19]. There are four stages of degradation, namely (i) hydration, (ii) initial degradation, (iii) constant degradation, and (iv) solubilization, as shown in Figure 1. Initially, water penetrates the polymeric matrix, disrupting van der Waals forces and hydrogen bonds, leading to a decrease in the glass transition temperature (Tg). In the initial degradation stage, covalent bonds are cleaved, reducing molecular weights. This is followed by constant degradation, characterized by autocatalysis of the carboxylic end cap and significant cleavage of backbone covalent bonds, leading to mass and integrity loss. Finally, in the solubilization stage, the polymer fragments are cleaved into smaller molecules, allowing the PLGA to dissolve in the aqueous environment [20]. Lactic acid and glycolic acid are considered the products of PLGA biodegradation and are eliminated from the body through the Krebs cycle. Lactic acid is converted into carbon dioxide and pyruvate, which enter the Krebs cycle, while glycolic acid is either excreted in urine or oxidized to glyoxylate, which is also converted into pyruvate and further processed in the Krebs cycle. This results in the production of carbon dioxide and water, which are safely secreted, minimizing systemic toxicity [21,22,23].
Various biodegradable devices and pharmaceutical drug delivery products utilize PLGA, including microparticles (MPs), nanoparticles (NPs), implants, and micelles [1]. MPs, which include spherical microspheres, are larger particles made from various materials and range in size from 1 to 1000 micrometers. They are commonly used for drug delivery, tissue engineering, and other applications [24]. NPs, on the other hand, are small particles in the nanometer range with various shapes, such as rods, cubes, and tubes [25]. Nanofibers, a type of NP, are elongated, thin fibers, while nanospheres are spherical nanoparticles with a high surface area-to-volume ratio, allowing for effective drug loading [26]. Implants are devices inserted into the body and commonly used in orthopedics, cardiology, and other medical fields for sustained drug delivery or structural support [27]. Micelles are self-assembling structures composed of amphiphilic molecules and are commonly used to encapsulate and solubilize hydrophobic drugs within the core and release them in a controlled manner [28].
PLGA serves as both a drug carrier and as the structural framework for drug-loaded materials [29]. In recent decades, the Food and Drug Administration (FDA) has approved many drug delivery products that have become some of the most successful complex drugs on the market. PLGA-based drug carriers have been particularly effective due to their high encapsulation efficiency, superior biocompatibility, stable release performance, and sufficient bioavailability [16]. Additionally, PLGA-based DDSs, particularly those using nano-PLGA, enhance aqueous solubility, bioavailability, and the stability of APIs [30].
Currently, PLGA-based DDSs are being developed to deliver small-molecule drugs, vaccines, proteins, and peptides. Since the introduction of the Lupron Depot® (leuprolide) in 1989, the FDA has approved several PLGA-based drug products, including Risperdal Consta® (risperidone) and the peptide-containing microspheres of Lupron Depot® (leuprolide), as shown in Table 1 [31,32]. Moreover, clinical trials have shown promising results by exploring new indications for existing products or investigating potential new therapeutics. Several studies [33,34] have summarized the various phases of ongoing PLGA-based clinical trials registered at ClinicalTrails.gov [35]. For example, a study on naltrexone-loaded microspheres (marketed as Vivitrol®) showed approximately 33.70 wt% drug loading and 66.30% of the formulation containing 75:25 PLGA in each vial [36]. In comparison, the commercial Lupron Depot® formulation has a leuprolide acetate loading of 10% and a gelatin loading of 1.7%, with the aliphatic ester end-capped and a 75:25 ratio of PLGA [37].
In recent decades, significant advancements in the continued research and development of PLGA-based formulation technologies have demonstrated innovation and potential to improve therapeutic outcomes and address complex healthcare challenges. This work discusses the latest research on PLGA applications. PLGA nanoparticles can improve drug stability, enhance drug targeting, and provide controlled release profiles for drug delivery applications. Innovative methods for PLGA nanoparticle formulation, such as emulsion solvent evaporation, nanoprecipitation, and electrospraying, are being explored and have shown outstanding outcomes [38,39]. Additionally, stimuli-responsive PLGA formulations that respond to specific triggers, such as changes in pH and temperature, have been investigated to reduce side effects and enhance the therapeutic efficacy of drug targeting [40,41,42,43]. The development of PLGA-based delivery systems for combination therapy, which co-encapsulates multiple drugs or therapeutic agents in a single formulation, is another research focus aimed at improving treatment outcomes by targeting multiple pathways or addressing different aspects of a disease simultaneously [44]. Researchers are also exploring the development of hybrid DDSs that combine PLGA with other polymers or biomaterials, such as PEG and chitosan, to enhance the targeted properties and drug delivery performance of the formulations [45]. These hybrid systems offer synergistic effects for the delivery of drugs to the target sites, such as the brain, improving biocompatibility and tailoring drug release profiles. This approach has the potential to reduce drug resistance and improve treatment outcomes [46,47].
Figure 1. The synthesis and degradation of PLGA. (A) The ROP of lactide and glycolide. Source: [17]. (B) Direct melt condensation polymerization. Source: [15]. (C,D) Schematic diagram of PLGA in different degradation stages. Source: [48]. (E) The chemical structure of PLGA with different end caps (the asterisk indicates S,S stereoisomer).
Figure 1. The synthesis and degradation of PLGA. (A) The ROP of lactide and glycolide. Source: [17]. (B) Direct melt condensation polymerization. Source: [15]. (C,D) Schematic diagram of PLGA in different degradation stages. Source: [48]. (E) The chemical structure of PLGA with different end caps (the asterisk indicates S,S stereoisomer).
Polymers 16 02606 g001

2. Physicochemical Properties of PLGA

PLGA, a copolymer of lactic acid and glycolic acid, is synthesized through ring-opening polymerization [17,49] of the corresponding lactic acid dimers and glycolic acid dimers or by direct polycondensation of lactic acid and glycolic acid. Upon degradation, it breaks down into carbon dioxide and water, as illustrated in Figure 1 [19]. These copolymers are available with various key properties, including molecular weight, intrinsic viscosity, monomer ratio (L: G ratio), blockiness, and end caps, all of which influence degradation periods ranging from months to years. The critical quality attributes of drug delivery formulations, such as particle size (or distribution) and porosity, exhibit significant differences that affect both in vitro and in vivo performance [50]. It is well known that PLGA characteristics such as a higher molecular weight [51], a higher monomer ratio [52], lower blockiness [53], and acid end caps [54,55] contribute to lower hydrophilicity and, consequently, slower drug release. For example, according to the labeling information approved by the FDA, Leuprolide acetate for injectable suspension, marketed under the brand name ELIGARD®, is formulated using different copolymers with varying end caps and monomer ratios. ELIGARD® 7.5 mg is composed of 7.5 mg of drug loading and 82.5 mg of a 50:50 copolymer containing a carboxyl end cap, whereas ELIGARD® 22.5 mg includes 22.5 mg of drug loading and 158.6 mg of a 75:25 copolymer with hexanediol.

2.1. Molecular Weight

PLGA exhibits a range of molecular weights, spanning from under 10,000 to up to 200,000 g/mol [56]. These molecular weights can be determined using intrinsic viscosity measurements or gel permeation chromatography (GPC) with polystyrene external standards (PESs). However, due to differences in polymer–solvent interactions, PLGA’s molecular dimensions differ from those of polystyrene, leading to potential inaccuracies when using polystyrene standards as a reference. In commercial practice, intrinsic viscosity is frequently employed to estimate the molecular weights of PLGAs. However, this method’s accuracy is limited by the complex relationship between PLGA’s molecular dimensions and the monomer ratio. Consequently, estimating the molecular weights of PLGAs with varying L/G ratios based solely on a standard curve can be challenging [57]. For improved accuracy, multi-angle static light scattering (MALS) is utilized to measure the molecular weights of PLGAs. Compared with the molecular weights obtained via MALS, those obtained by the PES method may differ by up to 70% under certain conditions [57,58]. As the degradation rate increases due to hydrolysis, lower-molecular-weight PLGA is less likely to exhibit hydrophobic characteristics [59]. The molecular weight of PLGA plays a crucial role in determining the drug release kinetics, mechanical properties, stability, and drug-loading capacity of PLGA-based DDSs. Low-molecular-weight PLGA polymers typically degrade more rapidly, resulting in accelerated drug release, often characterized by an initial burst release followed by a sustained release over time [60]. Additionally, low-molecular-weight PLGA polymers tend to have lower mechanical strength and stiffness, which can compromise structural integrity and stability, potentially leading to premature drug release or reduced drug efficacy. Furthermore, lower-molecular-weight polymers may offer reduced drug-loading capacity due to limited space for drug encapsulation, affecting overall drug delivery efficiency.

2.2. Intrinsic Viscosity

A crucial aspect of polymer molecular weight is the intrinsic viscosity of PLGA, which is directly correlated with particle size and drug release rate. High intrinsic viscosity in PLGA polymers, which is associated with high molecular weight, leads to larger particle sizes and a slow drug release rate [61]. Additionally, increased intrinsic viscosity reduces the tendency of protein molecules to diffuse through the polymer matrix, thereby enhancing the encapsulation efficiency of proteins/peptides [62]. The higher intrinsic viscosity also imparts greater mechanical strength and stiffness, which contribute to the structural integrity of the DDS, resulting in improved stability over time and a longer shelf life of DDSs [63].

2.3. Monomer Ratio

The ratio of glycolide and lactide impacts the polymerization and properties of PLGA [64]. For instance, PLGA with a 40:60 ratio indicates that the polymer is composed of 40% lactic acid and 60% glycolic acid. Commonly used PLGA monomer ratios are 50:50 and 75:25 [65]. Due to the higher hydrophilicity of glycolic acid compared to lactic acid, PLGA with a greater glycolic acid content exhibits faster hydrolysis and degradation rates, resulting in a shorter degradation time [66]. As illustrated in Figure 2, degradation decreases with a higher L: G ratio, as the more hydrophobic methyl group leads to gentler absorption and slower water diffusion [57]. The monomer ratio greatly influences the properties of PLGA and is crucial in determining its performance in drug delivery applications. Generally, increasing the glycolic acid content in PLGA results in a more acidic microenvironment and faster degradation due to the higher hydrophilicity of glycolic acid relative to lactic acid. This change accelerates drug release from the delivery system [67,68]. By adjusting the monomer ratio, it is possible to customize the drug release profile to achieve sustained or controlled release of the drug over a desired period. Compared to PLGA with a higher lactic acid content, PLGA with a higher glycolic acid content tends to be more brittle and has lower tensile strength [69]. Therefore, the choice of monomer ratio is critical for applications where mechanical properties are important, such as in implantable devices. Careful selection of the monomer ratio allows researchers to tailor the properties of PLGA-based delivery systems to meet specific therapeutic requirements and improve their efficacy and safety.

2.4. Blockiness

The blockiness of PLGA refers to the alternating sequence of lactic acid and glycolic acid units within the polymer chain. This blocky structure imparts unique properties to PLGA, including tunable degradation rates, mechanical strength, and biocompatibility [70].
Research [71,72] indicates that one of the primary mechanisms of drug release from PLGA-based microspheres is hydrolytic degradation in aqueous PLGA media. Studies on microsphere degradation [53] have shown that the molecular weight distribution of PLGA with higher blockiness changes more significantly over time. This is because polymers with higher blockiness exhibit greater cleavage of the G-G linkages in aqueous environments compared to those with lower blockiness. Additionally, the blockiness of PLGA affects the stability of PLGA-based DDSs by influencing resistance to hydrolysis. Block copolymers with well-defined blocks of lactic acid and glycolic acid units generally exhibit greater stability than random copolymers. Due to their heterogeneous distribution of monomer units, random copolymers tend to degrade more quickly, leading to a faster drug release from PLGA-based DDSs [73]. Furthermore, the arrangement of lactic acid and glycolic acid units within the polymer chain affects drug interactions, thereby influencing the drug-loading efficiency and capacity of PLGA-based DDSs [74]. Consequently, blockiness is a crucial factor in designing and optimizing PLGA-based DDSs for specific therapeutic applications.

2.5. End Caps

Commercial PLGA typically features two main types of end caps, namely acid end caps and ester end caps, as illustrated in Figure 1. Ester-capped PLGA includes a long alkyl chain, often utilizing decanol or dodecanol as the initiator. In contrast, acid end-capped PLGA is produced by using lactic acid as an initiator and lacks this long alkyl chain [75]. The primary distinction between these two types of end caps is the presence of a long alkyl chain with a terminal methyl in the ester end-caped PLGA. This can be confirmed by detecting a methyl signal at 14 ppm in the 13C NMR spectrum for ester end-capped PLGA, while the absence of this peak indicates an acid end cap [75,76].
The choice of end cap influences the characteristics of PLGA microspheres. PLGA with ester end caps generally degrades more slowly compared to those with acid end caps [21]. In addition, there is a positive correlation between the presence of acid end caps and the release rate of hydrophobic drugs [77]. The acid end cap facilitates swelling of the PLGA matrix by initiating hydrolysis due to its water uptake potential. Consequently, among PLGAs with similar molecular weights and L:G ratios, ester end-capped PLGA typically exhibits a delay of four to six weeks in degradation in vivo compared to acid end-capped PLGA [78]. In recent decades, PLGA has been synthesized and modified with various functional groups to create novel end-capped copolymers. These advancements have expanded the applications of PLGA-based DDSs for a variety of purposes [17,18,79,80,81], as depicted in Figure 1.

3. Modification of PLGA-Based DDSs

To date, PLGA has been extensively modified to enhance its properties for DDSs. Researchers can choose from a range of modification strategies, depending on the desired outcomes and specific requirements of the delivery system.
Several approaches can be employed to modify PLGA, including surface modification, coating, blending, functionalization, and cross linking. Surface modification can be achieved by conjugating functional groups, such as amino groups or carboxylic acids, to the surface of PLGA nanoparticles. These functional groups enable the attachment of targeting ligands, antibodies, or imaging agents, thereby improving the stability, biocompatibility, or targeting capabilities of DDSs [82]. Coating with other polymers or materials can also modify PLGA surface properties. For instance, coating with PEG can extend the circulation time of PLGA nanoparticles in the body and reduce the immune response. Coating with chitosan or alginate, on the other hand, can enhance the mucoadhesive properties of PLGA, making it suitable for targeted delivery to mucosal surfaces [83]. Blending PLGA with other polymers allows for the creation of hybrid materials with tailored properties. For example, blending PLGA with polyethylene oxide (PEO) can improve the flexibility and mechanical strength of the polymer [84], while blending with PEG can enhance its hydrophilicity and drug release profile [85]. Chemical functionalization of PLGA introduces specific functional groups that can be used for drug conjugation, targeting, or controlled release. For example, functionalizing PLGA with hydrophilic polymers, peptides, or proteins can improve its interaction with biological tissues [86]. Cross linking PLGA nanoparticles can enhance their mechanical properties and stability, which improves drug-loading capacity and enables sustained release profiles. An example is the use of dimethyl dioctadecyl ammonium bromidebe (DDAB) to cross link protein with PLGA, facilitating stable delivery of protein into mitochondria [87].
For example, self-assembly structures of micelles are formed by amphiphilic molecules of dextran with PLGA by covalent bonding. PLGA serves as the hydrophobic copolymer, while dextran forms the hydrophilic shell due to its variable linkages and natural branches. This naturally creates an amphiphilic block copolymer that self-assembles into micelles [88]. Another example involves aspirin-loaded PLGA nanofiber coatings applied to the surface of titanium via electrospinning. These coatings can release aspirin over a period of up to 60 days [89]. Moreover, they promote the proliferation and osteogenic differentiation of bone mesenchymal stem cells while inhibiting M1 polarization and osteoclast differentiation of macrophages in vitro, representing a potential therapy to prevent inflammation and periprosthetic bone loss.

4. Load of PLGA-Based DDSs

4.1. Small-Molecule Drugs

Risperidone is a small-molecule, atypical antipsychotic agent known for its water-insoluble properties and its use in the treatment of neurological disorders, particularly schizophrenia, where it causes fewer extrapyramidal side effects (EPSs) than other antipsychotics [90]. However, the incidence of EPSs is dose-dependent, making it necessary to administer lower doses over a long-term period for effective management of schizophrenia [91]. To achieve stable and sustained delivery, implantable formulations of risperidone using PLGA have been developed and tested both in vitro and in vivo for pharmacokinetic profiles. These implantable formulations provide stable and long-term delivery of atypical antipsychotics [27]. Research conducted by the team of Dr. Burgess [91] investigated how variations in PLGA characteristics influence the in vitro and in vivo outcomes of risperidone in situ-forming implants (e.g., different molecular weights (Mw), L:G ratios, and end caps). Their findings revealed that even a 5 kDa deviation in molecular Weight (Mw), a 5% shift of the L:G ratio, and differences in the end caps of PLGA altered the release behavior of risperidone in both in vitro and in vivo settings. These results underscore the importance of carefully monitoring and controlling critical quality attributes when developing risperidone in situ-forming implant products. Previous studies [92] successfully established and validated a level-A correlation for risperidone-loaded PLGA microspheres, demonstrating the potential of these formulations in achieving consistent and predictable drug release profiles. For instance, naltrexone, the API in Vivitrol®, is used to prepare PLGA microspheres, as it is a water-soluble compound that differs from risperidone. Combining a manufacturing process and dissimilar PLGAs, Vivitrol®, has been used to prepare PLGA microspheres due to its water-soluble nature, which contrasts with the water-insolubility of risperidone. The manufacturing process and different PLGA formulations used in creating Vivitrol® result in microspheres that exhibit biphasic release characteristics, showing a high in vitro–in vivo correlation (IVIVC) [93]. Once IVIVC is established, in vitro release methods can serve as surrogate models for bioequivalence studies, potentially reducing the need for extensive animal testing and minimizing the duration and cost of clinical trials [32]. This approach facilitates more efficient drug development processes, particularly for PLGA-based delivery systems designed to release water-soluble APIs with biphasic release profiles.

4.2. Natural Product

Paclitaxel (PTX) is a well-known taxane diterpenoid and the best-selling anticancer drug derived from natural sources, marketed under the brand name Taxol. It is widely used in the treatment of breast, lung, and ovarian cancers [94]. However, the clinical efficacy of PTX is limited due to its lipophilic nature, which results in low solubility, non-selective targeting, and rapid clearance from the body, leading to significant side effects [95]. To improve its solubility, formulations of PTX often include solubilized excipients such as Cremophor EL (CrEL) and absolute ethanol. However, these excipients are associated with severe side effects, including allergic reactions, nephrotoxicity, cardiotoxicity, etc. [96]. To address these limitations, paclitaxel-loaded PLGA nanospheres have been developed. These nanospheres utilize biodegradable polymers to provide a slower and sustained release of PTX, significantly enhancing its antitumor effect while reducing side effects compared to traditional formulations [97].
Another natural product, curcumin (CUR), is extracted from the dried rhizome of Curcuma longa L. and is widely recognized as a chemosensitizer in cancer therapy [98]. Combining CUR with PTX represents a promising cancer treatment strategy, enabling dosage reduction to minimize side effects while improving therapeutic efficacy. Several clinical studies, including the NCT01490996 project sponsored by the University of Leicester, are investigating the combination of CUR with chemotherapy in patients with inoperable colorectal cancer, with some trials currently in phases I and II [35]. However, both CUR and PTX suffer from poor water solubility and low bioavailability, which can limit their therapeutic effectiveness [99]. To overcome these challenges, nanocarriers are employed to encapsulate and protect CUR and PTX, thereby improving their solubility and chemical stability. This encapsulation strategy enhances the circulation time of these drugs in the body, ultimately improving their therapeutic outcomes [100].

4.3. Protein or Peptide

Proteins and peptide drugs (PPDs) are highly specific and potent therapeutic agents in pharmaceutical applications. The controlled delivery and release of these biologics have lots of benefits, including improved efficacy, enhanced accessibility, mitigation of dosing side effects, increased patient convenience, and better adherence to treatment regimens [101]. Various techniques have been developed to prepare nanoparticles for loading with PPDs, such as protein adsorption [102] and coating techniques. These methods open up the possibility of introducing biological ligands into the coatings of peptides and anti-protein microspheres in the future, thereby promoting preferential interaction and uptake into the targeted cells. Microspheres with low-protein-binding surfaces that expose specific biological functions provide a versatile platform for the design of targeted drug delivery systems [103].
For instance, leuprolide acetate PLGA MPs are commonly used in the treatment of various medical conditions, such as prostate cancer, endometriosis, and certain types of infertility. These PLGA MPs are injected at the site of action, where they slowly degrade over time, releasing the drug in a controlled manner. This approach has proven effective in managing symptoms and improving the quality of life of patients with these conditions. In a study by Pro. Burgess [104], after comparison with leuprolide acetate MP formulations with varying physicochemical properties, drug release appeared to be dependent on the molecular weight and the weight distribution of PLGA. Specifically, the initial burst release of the drug increased with a higher proportion of low-molecular-weight PLGA, underscoring the importance of controlling burst release in these formulations.
Furthermore, proteins and peptides are often released from the PLGA matrix following a biphasic release profile characterized by an initial burst phase followed by a gradual drug release phase. This release pattern can be influenced by the interactions between the polymers and the drugs, such as peptides with positive charges loaded into the negatively charged polymers or acylated peptides that from adducts during degradation, which exhibit slow release properties over extended periods [33].

4.4. Antibiotics or Antiviral Drugs

Antigen-presenting cells beneath the epidermis can be effectively targeted using mannose-modified PLGA nanoparticles (MNPs) loaded with the hepatitis B surface antigen (HBsAg) protein. This approach has been shown to induce potent adaptive immunity responses by enhancing antigen uptake by immune cells [105]. Due to the properties of PLGA, long-lasting immunity can be established through the slow release of HBsAg, which allows for sustained presentation to lymphocytes. This strategy holds significant potential for future vaccine development, as illustrated in Figure 3. Furthermore, the van der Waals forces conferred by mannose modification increase the affinity for proteins, while glycosylated PLGA can improve antigen stability upon release. Overall, mannosylated PLGA NPs represent a promising research direction in terms of increasing delivery efficiency and specificity to targeted cells by inducing the receptor-mediated endocytosis of NPs.
Prodigiosin (PG) is a bioactive red pigment produced through microbial fermentation of Serratia marcescens strains, as well as other microorganisms. Being an effective proapoptotic agent against various cancer cells, including those with multidrug resistance cells, PG demonstrates its superiority with minimal toxicity on normal cells [106]. The delivery of PG to breast tumors using PLGA-based microparticles was studied with paclitaxel serving as a control [107]. The research demonstrated that PG-loaded PLGA microparticles, which release PG at controlled rates, significantly reduce the viability of MDA-MB-231 breast cancer cells by halting mitosis and inducing apoptosis. Additionally, a clinical study was initiated in 2021 (NCT04735601), investigating Ahmed valve implantation with mitomycin C-coated PLGA for the treatment of Sturge Weber syndrome glaucoma in adults. This study is in Phase III, although results have not yet been posted [35].

5. Applications of PLGA-Based DDSs

5.1. Pain

Severe side effects may result from repeated use of systemic administration of analgesic drugs. In contrast, persistent pain can be treated with or without reduced side effects using local anesthetics (LA; e.g., ropivacaine [RVC]). However, the effect of local anesthetics after a single injection lasts only several hours. Postoperative pain may last for a few days, several months, or even more than 1 year. Therefore, producing a prolonged analgesic effect of new LA formulations is a compelling need. Sustained-release RVC (SRR) and its effect on postsurgical and neuropathic pain can be observed using RVC-loaded PLGA particles. The administration of local SRR [108] has been shown to have a long-lasting antinociceptive effect on neuropathic pain with biosecurity and tolerance by the body.
The epidural space of the spine can be injected with an anti-inflammatory agent dexamethasone (DEX) and local anesthetic ropivacaine (RVC) in clinical settings to treat neuropathic pain in patients. However, uncontrolled flow of short-acting drugs to the motor nerve is observed in association with such drug treatment [8]. Therefore, PLGA nanofibers containing DEX and RVC (PGLA-CD-DEX-RVC nanofibers) have been developed to overcome these limitations. The nanofiber scaffold enables many drugs to onboard the surface. Furthermore, synthesized PLGA-CD-DEX-RVC nanofibers release both drugs sustainably for over 48 h and relieve allodynia cold sensitivity consistently for 14 days. Moreover, these nanofibers can prevents drugs from entering motor nerves, as shown in Figure 4. PLGA-CD-DEX-RVC nanofibers are a good longer-term treatment option for neuropathic pain in clinical settings.

5.2. Cancers

Photothermal therapy (PTT) is a treatment method that uses light-absorbing agents to produce heat upon exposure to laser light. Targeted heating can effectively pinpoint and eliminate cancer cells or other unhealthy tissues while preserving the integrity of surrounding healthy tissue. Utilizing PLGA as a carrier for light-absorbing agents (e.g., gold nanoparticles or organic dyes) allows for controlled release at the designated target site upon laser light activation and reduces systemic toxicity, making it a promising approach for the safe and effective long-term treatment of various diseases [109]. PLGA loaded with black phosphorus quantum dots (BPQDs) can produce biodegradable BPQD/PLGA nanospheres with oil-in-water emulsion solvent evaporation. The hydrophobic PLGA prevents their rapid degradation by isolating the interior BPQDs. After intravenous injection, the NSs boast long blood circulation in the body through the EPR effect, which enhances photothermal stability and efficient tumor aggregation for efficient photothermal cancer treatment. BPQDs/PLGA NSs can be degraded to nontoxic, biocompatible products and cleared in a certain period after completing the therapeutic functions [110]. A biotin-conjugated PLGA polymer for irinotecan nanoparticles (PLGA-B-NP-Ir), as shown in Figure 5, can improve the solubility of the drug after biotin conjugation and advance active targeting, as well as the EPR effect, leading to significantly delayed tumor growth after multiple systemic injections in a mouse model [17]. This suggests the potential for active targeted drug delivery applications for cancer treatment.
Autophagy inducers improve tumor-cell sensitivity to chemotherapeutic drugs and boost antitumor efficacy [111]. Thus, the use of a combination of autophagy inducers and anticancer drugs is a potential strategy for cancer therapy. According to the results of Zhang’s study, autophagosomes can be degraded into lysosomes by PLGA-based NPs, inducing autophagy; therefore, autophagy inhibitors were combined with drug-loaded NPs. The NPs from endosomes and lysosomes causing NPs can be degraded by autophagy inhibitors to sustain existing drug release in the cytoplasm. In addition, autophagy inhibitors can block the autophagy from cancer cells to resist chemotherapeutic drugs [112]. Docetaxel with PLGA NPs, together with autophagy inhibitors (e.g., 3-MA and CQ), can greatly boost the both in vitro and in vivo therapeutic outcomes [113].
Surface modification with PEG can shift PLGA-based NPs with negative charges to neutral or positive values [114]. PLGA NPs endowed with the ability to escape from the endo-lysosome have many potential applications because lysosomes are the drug target, although they should be avoided for the degradation of PLGA-based NPs and more efficient drug delivery [115]. Furthermore, surface modification can enhance the biocompatibility of hydrophobic PLGA NPs. Such a modification can be performed by hydrophilic molecules, such as PEG and d-a-tocopheryl polyethylene glycol 1000 succinate (a water-soluble derivative of natural vitamin E and TPGS). This may be because PEG and TPGS-modified NPs are more easily recognized by and induce autophagosomes [113]. Additionally, the pre-existence of cancer-specific cytotoxic T lymphocytes (CTL) at the tumor site can be achieved by effectively treating immune checkpoint blockade. The authors of [116] studied PLGA particle-elicited CTL responses to multiple human tumor peptide antigens of various cancers.

5.3. Neurological Disorders

Pharmacotherapy for central nervous system disorders is complicated, as the BBB and extensive first-pass metabolism impede most chemical and biological drugs. Thus, it leads to low therapeutic efficacy and aggravated side effects caused by dose increase and drug accumulation [117]. Adsorptive-mediated transcytosis (AMT) provides a pathway for drug transport to the brain targeted across the BBB [118]. A mannose coating over PLGA NPs for delivery of Donepezil (DPZ) and Memantine (MEM) holds excellent prospects for the treatment of Alzheimer’s disease. Mannose-coated PLGA NPs are targeted for effective brain delivery [105], as ligand coating provides improved targeting with AMT [119], as applied in copolamine-induced a murine model via the intranasal (IN) route [120]. This could ferry biologic drugs effectively to the brain by overcoming the drawbacks of oral and intravenous routes [121]. Mannose-coated PLGA NPs not only provide excellent targeting and improved brain delivery but also offer maximum bioavailability of both drugs in the brain and minimize exposure to peripheral organs [120].
Curcumin, a hydrophobic polyphenol medication known for its remarkable pharmacological effects on the nervous system, potentially alleviates neurological disorders [122]. However, the clinical application of curcumin is hampered by various factors, such as poor solubility, low permeability, low stability in the body fluids under low aqueous solubility, limited bioavailability, rapid clearance, and reduced absorption in the gastrointestinal (GI) tract [123,124]. PLGA NP-based delivery systems enhance the aqueous solubility and bioavailability of hydrophobic curcumin, enabling it to penetrate the BBB and improving its distribution throughout the body. Furthermore, the encapsulation of curcumin in NPs is believed to enhance its stability and protect it from enzymatic and pH degradation [98,122,125].
Consequently, PLGA-based DDSs provide a consistent and convenient medication regimen to help improve neurological patient compliance, leading to better treatment outcomes and improved overall quality of life.
Microcapsules with gelatin methacrylate cores and PLGA shells (GelMa-PLGA MPs) are designed using a capillary microfluidic technique for encapsulated ginkgo biloba extract (GBE) delivery, as shown in Figure 6, as the low bioavailability and short half-life of GBE in vivo limit their neurological application. GBE is slowly released from solidified microcapsules during PLGA degradation and shows good biocompatibility in cell experiments. When GBE-containing GelMa-PLGA MPs are applied to APP/PS1 mice, amyloid deposition is reduced in the mouse brain, and cognitive impairment is improved. These results indicate that GBE-encapsulated MPs have potential clinical value in treating AD and other neurodegenerative diseases [126].

5.4. Inflammation

Osteoarthritis (OA) is the most common chronic joint disorder caused by degeneration or loss of articular cartilage. However, articular cartilage damage that occurs during OA progression cannot be treated. Tetramethylpyrazine (TMP) has strong anti-inflammatory and chondroprotective activities but is rapidly cleared from the joint cavity after joint injection. Thus, multiple injections are required to maintain the efficacy of treatment. Encapsulation of TMP in PLGA microspheres enhanced TMP retention within the joint for 30 days, reduced the number of injections, and decreased dosage from 12.6 mg to 4.6 mg compared with a TMP solution. Joint injection of TMP microspheres efficiently relieves inflammatory symptoms, improves joint lesions, and decreases chondrocyte depletion [127].
Figure 6. Examples of PLGA applications for neurological disorders. (A) Schematic capillary microfluidic chip for preparation of GBE-GelMa-PLGA MPs in double-emulsion droplets. (B) Illustration of MP degradation and drug release. (C) Search paths of mice in different groups of the Morris water maze (MWM) test. (D) Fluorescence images of N2a cells in the control group (blank well) and the experimental group (GelMa-PLGA MPs) taken using a fluorescence microscope. Source: [126].
Figure 6. Examples of PLGA applications for neurological disorders. (A) Schematic capillary microfluidic chip for preparation of GBE-GelMa-PLGA MPs in double-emulsion droplets. (B) Illustration of MP degradation and drug release. (C) Search paths of mice in different groups of the Morris water maze (MWM) test. (D) Fluorescence images of N2a cells in the control group (blank well) and the experimental group (GelMa-PLGA MPs) taken using a fluorescence microscope. Source: [126].
Polymers 16 02606 g006
Dictamnine (i.e., dictamnus dasycarpus) is an active pharmaceutical ingredient in Chinese herbal medicine that is commonly used to treat skin inflammation like atopic dermatitis. PLGA has been used as encapsulated dictamnine drug nanocarrier (Dic-PLGA-NC) to study anti-inflammatory mechanisms. Compared to naked dictamnine, Dic-PLGA-NC was found to improve the drug release rate and penetrate more efficiently into epidermal structures of a dermatitis mouse model induced by oxazolone, as shown in Figure 7, which may be attributed to the controlled, sustained release from PLGA carriers [128].
A monocyte membrane-coated, PLGA nanoparticle-based, gliclazide-loaded nanoghost was developed to control inflammation and atherosclerosis [129]. A nanoghost (NG) is a novel bioinspired, nanoscale vehicle with an increased half-life that escapes the immune response through cell membrane-coated surface functionalization.

5.5. Vaccines

In recent years, drug delivery technologies have been developed to improve human health in multiple ways, including by enhancing therapeutic delivery to target sites, minimizing off-target accumulation, and improving patient compliance [130]. Compared to traditional prime-boost regimes, single-dosing vaccines offer significant benefits with increasing vaccine population coverage, reducing costs and saving time, as patients only require one health check [131]. Furthermore, a single-dosing vaccine can rapidly induce a protective immune response, which is preferred under pandemic conditions. Still, many vaccines require more than one dose to fully protect the host at present [132].
A single-dose influenza vaccine derived from recombinant outer membrane vesicles (rOMVs) was designed as PLGA microparticles displaying an antigen-mapped heterospecies tandem sequence of influenza virus M2 protein for release over a month. The protective immune response is long-standing and elicited sustained antibody titers for six months after initial vaccination and 100% survival of mice severely infected with influenza [132]. Antigens and adjuvants encapsulated in PLGA lead to increased cellular response, partly because of enhanced antigen presentation by macrophages and dendritic cells (DCs) [133]. As the ability of dendritic cells to engulf depends on the particle size [134], determination of the ideal size is necessary in future works involving rOMVs within PLGA MPs in order to achieve a long-lasting, optimized, and potent cellular response. A multi-compound particulate vaccine targeting DCs was formulated as PLGA-based NPs to transport Ag protein and adjuvants via type I transmembrane protein CD40, intending to stimulate efficient cytotoxic CD8+ T-cell responses [135].
Figure 7. Examples of PLGA applications for the treatment of inflammation. (A) Schematic representation of the manufacturing process and animal model of Dic-PLGA-NC. (B) Representative confocal microscopic images showing cellular localization of endogenous p65 in oxazolone-stimulated HaCaT cells (arrows) with dicatmnine treatment. DD, dictamnus dasycarpus. Scale bar: 25 μm. (C) Representative images showing two-photon microscopic evaluation of mouse skin; this demonstrates that the nanoformulated dictamnine reached the dermal tissue layer and possessed superior skin penetration capability for AD therapy relative to naked dictamnine. (D) Dic-PLGA-NC cumulatively released over 216 h in PBS [128]. Copyright (2021), Elsevier.
Figure 7. Examples of PLGA applications for the treatment of inflammation. (A) Schematic representation of the manufacturing process and animal model of Dic-PLGA-NC. (B) Representative confocal microscopic images showing cellular localization of endogenous p65 in oxazolone-stimulated HaCaT cells (arrows) with dicatmnine treatment. DD, dictamnus dasycarpus. Scale bar: 25 μm. (C) Representative images showing two-photon microscopic evaluation of mouse skin; this demonstrates that the nanoformulated dictamnine reached the dermal tissue layer and possessed superior skin penetration capability for AD therapy relative to naked dictamnine. (D) Dic-PLGA-NC cumulatively released over 216 h in PBS [128]. Copyright (2021), Elsevier.
Polymers 16 02606 g007
Restricted by it limited bioavailability and poor immunogenicity, subunit antigen vaccination is unstably successful, and soluble antigens are often unsuccessful when cross-presented. DDSs formulated with PLGA can overcome these problems by protecting antigens from degradation and increasing biodistribution to improve the ability to be uptaken by antigen-presenting cells (APCs) [136].
Using a 3D manufacturing process, a transdermal PLGA core-shell microneedle patch was developed by tailoring PLGA degradation to precisely control drug release. The PLGA cores and shells enable the release of the vaccine payload in pre-programmed bursts over several days to over a month in a single administration. In the clinical use setting, multiple PLGA core shells with varying degradability kinetics settings in microneedles use skin a single injection for several burst releases across multiple periods. In rats, the use of Prevnar-13 in loaded microneedles for the bacterium Streptococcus pneumoniae caused similar immune responses as those to the bacterium Streptococcus pneumoniae with multiple bolus injections, as shown in Figure 8. Thus, pre-programmed microneedle patches offer alternative strategies for prevention and treatment regimens requiring multiple injections [137]. Such strategies may overcome the scalability and cost-effectiveness issues associated with producing large quantities of PLGA-based vaccines at a reasonable cost.

5.6. Tissue Regeneration

In regenerative medicine, PLGA has received recognition as a biological substitute, since the FDA approved its clinical use for restoring, maintaining, or improving damaged tissues [22]. PLGAs have been developed as regenerative medicine delivery carriers in some tissue regeneration and remodeling fields, such as skin, including wound healing, articular cartilage, bone regeneration, and muscle and neural remodeling [138,139,140,141,142]. However, long-term safety, encapsulation efficacy, inflammation, and immune reactions should be considered during regeneration treatment. Furthermore, 3D printing technology has been increasingly used to fabricate complex PLGA scaffolds that can mimic the structure and properties of native tissues, providing a platform for cell growth and tissue regeneration.
A unique combination of two minimally invasive therapy options involves the injection of modular microtissues. These microtissues consist of mouse myoblast (C2C12)-laden PLGA porous microspheres (PLGA PMs) and human umbilical vein endothelial cell (HUVEC)-laden PEG hollow microrods (PEG HMs), along with an established extracellular matrix (ECM) to facilitate the development of skeletal muscles in their natural locations. This highly porous structure of PLGA PMs can be applied to C2C12 cells to obtain an effective culture. Discrete C2C12-laden PLGA PMs can be constructed in 7 days of incubation, with strong cell–cell co-relation among C2C12 cells, resulting in the connection of the discrete PLGA PMs (denoted with red arrow in the CLSM image in Figure 9). Moreover, vascular analog-laden PEG HMs in muscle-like constructs have shown promising results in stimulating the enhancement of blood vessels after injection and improving newborn vessels [141].
A synthesized glycine–histidine–lysine peptide (GHK) conjugate with L-carnitine was loaded into PLGA NPs to improve wound healing and exert anti-inflammation effects. Even though GHK and L-carnitine play a crucial role in wound tissue regeneration, upon PLGA degradation, a lactic acid pool could accelerate wound closure in synergetic relation [143,144].

6. Limitations and Challenges of Using PLGA-Based DDSs

Even though PLGA is generally considered biocompatible, some studies suggest that PLGA may be toxic both in vitro and in vivo because of the presence of residual stabilizing molecules, inconsistent preparations, and batch-to-batch variations, potentially causing adverse effects on tissue health and environmental impacts [145]. Compared to simpler DDSs, PLGA formulations are more complex, requiring additional manufacturing steps and quality control measures, which result in higher manufacturing material costs. On the other hand, long-term PLGA-based DDSs can contribute to their cost savings by reducing the need for device replacement and leading to prolonged therapeutic effects, potentially lowering overall treatment costs. Considering the complexity and cost of production of PLGA-based DDSs on a large scale, ensuring consistent quality and reproducibility across batches is essential for clinical use. PLGA not only exhibits burst release with inconsistent drug release profiles, which may result in an initial overdose followed by a rapid decrease in drug concentration [146], but also a lower drug-loading capacity and stability, which restrict the drugs that can be delivered [147]. It is necessary to ensure the safety of PLGA and its effective drug delivery, including by enhancing the drug-loading capacity and the effect of encapsulation, which is one of the big challenges in developing PLGA-based DDSs. Acidic byproducts degraded from PLGA may also cause tissue irritation and accelerate drug release kinetics, making it difficult to achieve consistent and predictable drug release profiles in clinical settings. Despite extensive preclinical studies, there is still a limited understanding of the in vivo behavior of PLGA-based DDSs, including their pharmacokinetics and biodistribution. Moreover, the lack of standardized methods for evaluating the performance of PLGA-based DDSs is another main challenge, as PLGA is sensitive and can be influenced by environmental factors and its physicochemical properties. It is difficult but important to establish a stable IVIVC drug delivery system for consistent API performance and the development of product formulations. Optimizing novel polymer blends, developing smart stimuli-responsive systems, and investigating advanced drug delivery strategies (e.g., nanotechnology and 3D printing) can potentially further improve PLGA-based DDSs by enhancing drug-loading capacity and control drug release kinetics.

7. Conclusions and Future Perspectives

In conclusion, PLGA has emerged as a versatile and promising material for DDSs due to its biodegradability, biocompatibility, and tunable physicochemical properties. Using PLGA in the formulation of drug delivery pharmaceutical products, such as in situ-forming systems, nanoparticles, microspheres, and micelles, has shown significant potential in enhancing the efficacy of treatments and reducing the toxicity of various drugs. This review highlights recent research applications of PLGA to deliver small-molecule drugs, proteins, peptides, and antibiotics or antiviral agents. The applications of PLGA-based DDSs extend across a broad spectrum of diseases, including cancer, neurological disorders, pain, inflammation, vaccines, and tissue regeneration.
Future research should focus on optimizing the physicochemical properties of PLGA and exploring innovative strategies for targeted and controlled drug release to expand the clinical potential of PLGA-based DDSs further. Firstly, the development of PLGA in sustained-release systems for pain relief medications is both necessary and urgent, particularly in the context of rising concerns over drug abuse [148]. To achieve successful LADDS products, it is crucial to address challenges such as the initial burst release and subsequent incomplete release. Secondly, while novel PLGA NP-based vaccine technologies have become a research trend, they require further refinement, particularly in increasing drug load concentrations for human participants. This could lead to the development of long-acting vaccine delivery systems that provide targeted long-term immunotherapy for cancer treatment [149]. However, there still are various challenges that need to be solved, including the need for real-time monitoring of the in vivo fate to ensure the safety and ability of NPs to carry smaller genes for targeted delivery. Thirdly, surface-modified PLGA scaffolds hold promise for the loading of stem cells and immune cells for applications in tissue generation [150], inflammation, and neurological treatments [151]. Given the complexity of formulation development, it is necessary to establish effective IVIVC models to minimize time and costs and reduce reliance on animal and human studies. This involves the detailed exploration of the physicochemical properties that influence PLGA degradation rates, the development of predictive in vitro release tests, and reliable in vivo pharmacokinetic studies.

Author Contributions

J.Y.: literature review and writing—original draft. H.Z. and Y.L.: visualization and writing—original draft. Y.C. and M.W.: writing—review and editing. C.W.: supervision. P.H.: conceptualization, supervision, and writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (No. 82071367), and the Natural Science Foundation of Guangdong Province of China (Nos. 2021A1515220047 and 2023A1515011135).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. 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]
  2. Li, R.; Jia, Y.; Kong, X.; Nie, Y.; Deng, Y.; Liu, Y. Novel drug delivery systems and disease models for pulmonary fibrosis. J. Control. Release 2022, 348, 95–114. [Google Scholar] [CrossRef] [PubMed]
  3. Kashkooli, F.M.; Soltani, M.; Souri, M. Controlled anti-cancer drug release through advanced nano-drug delivery systems: Static and dynamic targeting strategies. J. Control. Release 2020, 327, 316–349. [Google Scholar] [CrossRef] [PubMed]
  4. Sabbagh, F.; Kim, B.S. Recent advances in polymeric transdermal drug delivery systems. J. Control. Release 2022, 341, 132–146. [Google Scholar] [CrossRef]
  5. Laracuente, M.L.; Marina, H.Y.; McHugh, K.J. Zero-order drug delivery: State of the art and future prospects. J. Control. Release 2020, 327, 834–856. [Google Scholar] [CrossRef]
  6. Shen, X.; Li, T.; Xie, X.; Feng, Y.; Chen, Z.; Yang, H.; Wu, C.; Deng, S.; Liu, Y. PLGA-based drug delivery systems for remotely triggered cancer therapeutic and diagnostic applications. Front. Bioeng. Biotechnol. 2020, 8, 381. [Google Scholar] [CrossRef]
  7. Rigon, L.; Salvalaio, M.; Pederzoli, F.; Legnini, E.; Duskey, J.T.; D’Avanzo, F.; De Filippis, C.; Ruozi, B.; Marin, O.; Vandelli, M.A.; et al. Targeting brain disease in MPSII: Preclinical evaluation of IDS-loaded PLGA nanoparticles. Int. J. Mol. Sci. 2019, 20, 2014. [Google Scholar] [CrossRef]
  8. Lee, D.; Nah, H.; Ko, W.K.; Kim, S.J.; Han, G.H.; Jeong, D.; Lee, D.; Han, I.; Sheen, S.H.; Heo, D.N.; et al. Thiolate poly (lactic-co-glycolic acid) nanofibers loaded with dexamethasone and ropivacaine show enhanced sustained release in the treatment of neuropathic pain through a local therapy technique. Chem. Eng. J. 2022, 431, 133356. [Google Scholar] [CrossRef]
  9. Deng, M.; Tan, J.; Hu, C.; Hou, T.; Peng, W.; Liu, J.; Yu, B.; Dai, Q.; Zhou, J.; Yang, Y.; et al. Modification of PLGA scaffold by MSC-derived extracellular matrix combats macrophage inflammation to initiate bone regeneration via TGF-β-induced protein. Adv. Healthc. Mater. 2020, 9, 2000353. [Google Scholar] [CrossRef]
  10. Gao, J.; Karp, J.M.; Langer, R.; Joshi, N. The future of drug delivery. Chem. Mater. 2023, 35, 359–363. [Google Scholar] [CrossRef]
  11. Wang, S.; Downing, G.; Olsen, K.F.; Sawyer, T.K.; Cone, R.D.; Schwendeman, S.P. Aqueous remote loading of setmelanotide in poly (lactic-co-glycolic acid) microspheres for long-term obesity treatment. J. Control. Release 2023, 364, 589–600. [Google Scholar] [CrossRef] [PubMed]
  12. Guarecuco, R.; Lu, J.; McHugh, K.J.; Norman, J.J.; Thapa, L.S.; Lydon, E.; Langer, R.; Jaklenec, A. Immunogenicity of pulsatile-release PLGA microspheres for single-injection vaccination. Vaccine 2018, 36, 3161–3168. [Google Scholar] [CrossRef]
  13. Li, W.; Tang, J.; Lee, D.; Tice, T.R.; Schwendeman, S.P.; Prausnitz, M.R. Clinical translation of long-acting drug delivery formulations. Nat. Rev. Mater. 2022, 7, 406–420. [Google Scholar] [CrossRef]
  14. 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]
  15. Gao, Q.; Lan, P.; Shao, H.; Hu, X. Direct synthesis with melt polycondensation and microstructure analysis of poly (L-lactic acid-co-glycolic acid). Polym. J. 2002, 34, 786–793. [Google Scholar] [CrossRef]
  16. Giram, P.S.; Wang, J.T.W.; Walters, A.A.; Rade, P.P.; Akhtar, M.; Han, S.; Faruqu, F.N.; Abdel-Bar, H.M.; Garnaik, B.; Al-Jamal, K.T. Green synthesis of methoxy-poly (ethylene glycol)-block-poly (L-lactide-co-glycolide) copolymer using zinc proline as a biocompatible initiator for irinotecan delivery to colon cancer in vivo. Biomater. Sci. 2021, 9, 795–806. [Google Scholar] [CrossRef]
  17. Giram, P.S.; Nimma, R.; Bulbule, A.; Yadav, A.S.; Gorain, M.; Venkata Radharani, N.N.; Kundu, G.C.; Garnaik, B. Poly (d, l-lactide-co-glycolide) Surface-Anchored Biotin-Loaded Irinotecan Nanoparticles for Active Targeting of Colon Cancer. ACS Omega 2024, 9, 3807–3826. [Google Scholar] [CrossRef]
  18. Rabanel, J.M.; Piec, P.A.; Landri, S.; Patten, S.A.; Ramassamy, C. Transport of PEGylated-PLA nanoparticles across a blood brain barrier model, entry into neuronal cells and in vivo brain bioavailability. J. Control. Release 2020, 328, 679–695. [Google Scholar] [CrossRef] [PubMed]
  19. Gentile, P.; Chiono, V.; Carmagnola, I.; Hatton, P.V. An overview of poly (lactic-co-glycolic) acid (PLGA)-based biomaterials for bone tissue engineering. Int. J. Mol. Sci. 2014, 15, 3640–3659. [Google Scholar] [CrossRef]
  20. Samir, A.; Ashour, F.H.; Hakim, A.A.; Bassyouni, M. Recent advances in biodegradable polymers for sustainable applications. NPJ Mater. Degrad. 2022, 6, 68. [Google Scholar] [CrossRef]
  21. Samadi, N.; Abbadessa, A.; Di Stefano, A.; Van Nostrum, C.; Vermonden, T.; Rahimian, S.; Teunissen, E.; Van Steenbergen, M.; Amidi, M.; Hennink, W. The effect of lauryl capping group on protein release and degradation of poly (d, l-lactic-co-glycolic acid) particles. J. Control. Release 2013, 172, 436–443. [Google Scholar] [CrossRef] [PubMed]
  22. Martins, C.; Sousa, F.; Araujo, F.; Sarmento, B. Functionalizing PLGA and PLGA derivatives for drug delivery and tissue regeneration applications. Adv. Healthc. Mater. 2018, 7, 1701035. [Google Scholar] [CrossRef]
  23. Zhao, D.; Zhu, T.; Li, J.; Cui, L.; Zhang, Z.; Zhuang, X.; Ding, J. Poly (lactic-co-glycolic acid)-based composite bone-substitute materials. Bioact. Mater. 2021, 6, 346–360. [Google Scholar] [CrossRef]
  24. Li, Q.; Chang, B.; Dong, H.; Liu, X. Functional microspheres for tissue regeneration. Bioact. Mater. 2023, 25, 485–499. [Google Scholar] [CrossRef]
  25. Behera, A. Nanomaterials. In Advanced Materials: An Introduction to Modern Materials Science; Springer International Publishing: Cham, Switzerland, 2022; pp. 77–125. [Google Scholar] [CrossRef]
  26. Mei, H.; Cai, S.; Huang, D.; Gao, H.; Cao, J.; He, B. Carrier-free nanodrugs with efficient drug delivery and release for cancer therapy: From intrinsic physicochemical properties to external modification. Bioact. Mater. 2022, 8, 220–240. [Google Scholar] [CrossRef]
  27. Magill, E.; Demartis, S.; Gavini, E.; Permana, A.D.; Thakur, R.R.S.; Adrianto, M.F.; Waite, D.; Glover, K.; Picco, C.J.; Korelidou, A.; et al. Solid implantable devices for sustained drug delivery. Adv. Drug Deliv. Rev. 2023, 199, 114950. [Google Scholar] [CrossRef] [PubMed]
  28. Zheng, Y.; Oz, Y.; Gu, Y.; Ahamad, N.; Shariati, K.; Chevalier, J.; Kapur, D.; Annabi, N. Rational design of polymeric micelles for targeted therapeutic delivery. Nano Today 2024, 55, 102147. [Google Scholar] [CrossRef]
  29. Loureiro, J.A.; Pereira, M.C. PLGA based drug carrier and pharmaceutical applications: The most recent advances. Pharmaceutics 2020, 12, 903. [Google Scholar] [CrossRef] [PubMed]
  30. Ding, D.; Zhu, Q. Recent advances of PLGA micro/nanoparticles for the delivery of biomacromolecular therapeutics. Mater. Sci. Eng. C 2018, 92, 1041–1060. [Google Scholar] [CrossRef]
  31. Wang, Y.; Qu, W.; Choi, S. FDA’s regulatory science program for generic PLA/PLGA-based drug products. Am. Pharm. Rev. 2016, 20. [Google Scholar]
  32. Wan, B.; Bao, Q.; Burgess, D. Long-acting PLGA microspheres: Advances in excipient and product analysis toward improved product understanding. Adv. Drug Deliv. Rev. 2023, 198, 114857. [Google Scholar] [CrossRef] [PubMed]
  33. Hu, F.; Qi, J.; Lu, Y.; He, H.; Wu, W. PLGA-based implants for sustained delivery of peptides/proteins: Current status, challenge and perspectives. Chin. Chem. Lett. 2023, 34, 108250. [Google Scholar] [CrossRef]
  34. 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]
  35. U.S. National Library of Medicine. Available online: https://www.clinicaltrials.gov/ (accessed on 8 June 2024).
  36. Hua, Y.; Wang, Z.; Wang, D.; Lin, X.; Liu, B.; Zhang, H.; Gao, J.; Zheng, A. Key factor study for generic long-acting PLGA microspheres based on a reverse engineering of Vivitrol®. Molecules 2021, 26, 1247. [Google Scholar] [CrossRef]
  37. Schutzman, R.; Shi, N.Q.; Olsen, K.F.; Ackermann, R.; Tang, J.; Liu, Y.Y.; Hong, J.K.; Wang, Y.; Qin, B.; Schwendeman, A.; et al. Mechanistic evaluation of the initial burst release of leuprolide from spray-dried PLGA microspheres. J. Control. Release 2023, 361, 297–313. [Google Scholar] [CrossRef]
  38. He, Y.; Chen, Q.W.; Yu, J.X.; Qin, S.Y.; Liu, W.L.; Ma, Y.H.; Chen, X.S.; Zhang, A.Q.; Zhang, X.Z.; Cheng, Y.J. Yeast cell membrane-camouflaged PLGA nanoparticle platform for enhanced cancer therapy. J. Control. Release 2023, 359, 347–358. [Google Scholar] [CrossRef] [PubMed]
  39. Kshirsagar, S.M.; Shrestha, N.; Kipping, T.; Banga, A.K. Formulation development of tazarotene-loaded PLGA nanoparticles for follicular delivery in the treatment of inflammatory skin diseases. Eur. J. Pharm. Biopharm. 2024, 200, 114346. [Google Scholar] [CrossRef]
  40. Palanikumar, L.; Al-Hosani, S.; Kalmouni, M.; Nguyen, V.P.; Ali, L.; Pasricha, R.; Barrera, F.N.; Magzoub, M. pH-responsive high stability polymeric nanoparticles for targeted delivery of anticancer therapeutics. Commun. Biol. 2020, 3, 95. [Google Scholar] [CrossRef]
  41. Zeeshan, M.; Ain, Q.U.; Weigmann, B.; Story, D.; Smith, B.R.; Ali, H. Dual pH and microbial-sensitive galactosylated polymeric nanocargoes for multi-level targeting to combat ulcerative colitis. Asian J. Pharm. Sci. 2023, 18, 100831. [Google Scholar] [CrossRef]
  42. Wei, D.; Sun, Y.; Zhu, H.; Fu, Q. Stimuli-responsive polymer-based nanosystems for cancer theranostics. ACS Nano 2023, 17, 23223–23261. [Google Scholar] [CrossRef]
  43. Braet, H.; Fransen, P.P.; Chen, Y.; Van Herck, S.; Mariën, R.; Vanhoorne, V.; Ceelen, W.; Madder, A.; Ballet, S.; Hoogenboom, R.; et al. Smart hydrogels delivered by high pressure aerosolization can prevent peritoneal adhesions. J. Control. Release 2023, 362, 138–150. [Google Scholar] [CrossRef] [PubMed]
  44. Lee, H.Y.; Kim, D.S.; Hwang, G.Y.; Lee, J.K.; Lee, H.L.; Jung, J.W.; Hwang, S.Y.; Baek, S.W.; lip Yoon, S.; Ha, Y.; et al. Multi-modulation of immune-inflammatory response using bioactive molecule-integrated PLGA composite for spinal fusion. Mater. Today Bio 2023, 19, 100611. [Google Scholar] [CrossRef]
  45. Watcharadulyarat, N.; Rattanatayarom, M.; Ruangsawasdi, N.; Patikarnmonthon, N. PEG–PLGA nanoparticles for encapsulating ciprofloxacin. Sci. Rep. 2023, 13, 266. [Google Scholar] [CrossRef] [PubMed]
  46. Giacon, N.; Lo Cascio, E.; Pennacchietti, V.; De Maio, F.; Santarelli, G.; Sibilia, D.; Tiberio, F.; Sanguinetti, M.; Lattanzi, W.; Toto, A.; et al. PDZ2-conjugated-PLGA nanoparticles are tiny heroes in the battle against SARS-CoV-2. Sci. Rep. 2024, 14, 13059. [Google Scholar] [CrossRef] [PubMed]
  47. Chauhan, M.; Shekhar, S.; Yadav, B.; Garg, V.; Dutt, R.; Mehata, A.K.; Goswami, P.; Koch, B.; Muthu, M.S.; Singh, R.P.; et al. AS1411 aptamer/RGD dual functionalized theranostic chitosan-PLGA nanoparticles for brain cancer treatment and imaging. Biomater. Adv. 2024, 160, 213833. [Google Scholar] [CrossRef]
  48. Hua, Y.; Su, Y.; Zhang, H.; Liu, N.; Wang, Z.; Gao, X.; Gao, J.; Zheng, A. Poly (lactic-co-glycolic acid) microsphere production based on quality by design: A review. Drug Deliv. 2021, 28, 1342–1355. [Google Scholar] [CrossRef]
  49. Pardeshi, S.R.; Nikam, A.; Chandak, P.; Mandale, V.; Naik, J.B.; Giram, P.S. Recent advances in PLGA based nanocarriers for drug delivery system: A state of the art review. Int. J. Polym. Mater. Polym. Biomater. 2023, 72, 49–78. [Google Scholar] [CrossRef]
  50. Wan, B.; Bao, Q.; Burgess, D.J. In vitro-in vivo correlation of PLGA microspheres: Effect of polymer source variation and temperature. J. Control. Release 2022, 347, 347–355. [Google Scholar] [CrossRef] [PubMed]
  51. Thakur, R.R.S.; McMillan, H.L.; Jones, D.S. Solvent induced phase inversion-based in situ forming controlled release drug delivery implants. J. Control. Release 2014, 176, 8–23. [Google Scholar] [CrossRef]
  52. Parent, M.; Nouvel, C.; Koerber, M.; Sapin, A.; Maincent, P.; Boudier, A. PLGA in situ implants formed by phase inversion: Critical physicochemical parameters to modulate drug release. J. Control. Release 2013, 172, 292–304. [Google Scholar] [CrossRef]
  53. Wan, B.; Bao, Q.; Zou, Y.; Wang, Y.; Burgess, D.J. Effect of polymer source variation on the properties and performance of risperidone microspheres. Int. J. Pharm. 2021, 610, 121265. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, X.; Burgess, D.J. Drug release from in situ forming implants and advances in release testing. Adv. Drug Deliv. Rev. 2021, 178, 113912. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, X.; Bao, Q.; Wang, R.; Wan, B.; Wang, Y.; Qin, B.; Burgess, D.J. Reverse engineering of Perseris and development of compositionally equivalent formulations. Int. J. Pharm. 2023, 639, 122948. [Google Scholar] [CrossRef] [PubMed]
  56. Sun, X.; Xu, C.; Wu, G.; Ye, Q.; Wang, C. Poly (lactic-co-glycolic acid): Applications and future prospects for periodontal tissue regeneration. Polymers 2017, 9, 189. [Google Scholar] [CrossRef]
  57. Park, K.; Skidmore, S.; Hadar, J.; Garner, J.; Park, H.; Otte, A.; Soh, B.K.; Yoon, G.; Yu, D.; Yun, Y.; et al. Injectable, long-acting PLGA formulations: Analyzing PLGA and understanding microparticle formation. J. Control. Release 2019, 304, 125–134. [Google Scholar] [CrossRef]
  58. Hadar, J.; Garner, J.; Skidmore, S.; Park, H.; Park, K.; Jhon, Y.; Wang, Y. Correlation analysis of refractive index (dn/dc) for PLGAs with different ratios of lactide to glycolide. In Proceedings of the 2018 Controlled Release Society (CRS) Annual Meeting, New York, NY, USA, 22–24 July 2018. [Google Scholar]
  59. Xu, Y.; Kim, C.S.; Saylor, D.M.; Koo, D. Polymer degradation and drug delivery in PLGA-based drug–polymer applications: A review of experiments and theories. J. Biomed. Mater. Res. Part B Appl. Biomater. 2017, 105, 1692–1716. [Google Scholar] [CrossRef]
  60. Smith, A.N.; Ulsh, J.B.; Gupta, R.; Tang, M.M.; Peredo, A.P.; Teinturier, T.D.; Mauck, R.L.; Gullbrand, S.; Hast, M.W. Characterization of degradation kinetics of additively manufactured PLGA under variable mechanical loading paradigms. J. Mech. Behav. Biomed. Mater. 2024, 153, 106457. [Google Scholar] [CrossRef]
  61. 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]
  62. Butreddy, A.; Gaddam, R.P.; Kommineni, N.; Dudhipala, N.; Voshavar, C. PLGA/PLA-based long-acting injectable depot microspheres in clinical use: Production and characterization overview for protein/peptide delivery. Int. J. Mol. Sci. 2021, 22, 8884. [Google Scholar] [CrossRef]
  63. Li, S.; Niu, D.; Shi, T.; Yun, W.; Yan, S.; Xu, G.; Yin, J. Injectable, in situ self-cross-linking, self-healing poly (l-glutamic acid)/polyethylene glycol hydrogels for cartilage tissue engineering. ACS Biomater. Sci. Eng. 2023, 9, 2625–2635. [Google Scholar] [CrossRef]
  64. Garner, J.; Skidmore, S.; Hadar, J.; Park, H.; Park, K.; Jhon, Y.K.; Qin, B.; Wang, Y. Analysis of semi-solvent effects for PLGA polymers. Int. J. Pharm. 2021, 602, 120627. [Google Scholar] [CrossRef] [PubMed]
  65. Astete, C.E.; Sabliov, C.M. Synthesis and characterization of PLGA nanoparticles. J. Biomater. Sci. Polym. Ed. 2006, 17, 247–289. [Google Scholar] [CrossRef]
  66. Wang, X.; Bao, Q.; Suh, M.S.; Kastellorizios, M.; Wang, R.; Burgess, D.J. Novel adapter method for in vitro release testing of in situ forming implants. Int. J. Pharm. 2022, 621, 121777. [Google Scholar] [CrossRef]
  67. Wang, Q.; Atluri, K.; Tiwari, A.K.; Babu, R.J. Exploring the application of micellar drug delivery systems in cancer nanomedicine. Pharmaceuticals 2023, 16, 433. [Google Scholar] [CrossRef]
  68. Kim, G.; Gavande, V.; Shaikh, V.; Lee, W.K. Degradation behavior of poly (lactide-co-glycolide) monolayers investigated by Langmuir technique: Accelerating Effect. Molecules 2023, 28, 4810. [Google Scholar] [CrossRef]
  69. Murcia Valderrama, M.A.; van Putten, R.J.; Gruter, G.J.M. PLGA barrier materials from CO2. The influence of lactide co-monomer on glycolic acid polyesters. ACS Appl. Polym. Mater. 2020, 2, 2706–2718. [Google Scholar] [CrossRef] [PubMed]
  70. 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]
  71. Avgoustakis, K. Polylactic-co-glycolic acid (PLGA). Encycl. Biomater. Biomed. Eng. 2005, 1, 1–11. [Google Scholar]
  72. 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]
  73. Patil, S.M.; Won, Y.Y. Effect of Monomer Sequence Distribution on the Glass Transition Temperature of Poly (d, l-lactic-co-glycolic acid) (PLGA). Macromolecules 2024, 57, 4947–4962. [Google Scholar] [CrossRef]
  74. De, R.; Mahata, M.K.; Kim, K.T. Structure-based varieties of polymeric nanocarriers and influences of their physicochemical properties on drug delivery profiles. Adv. Sci. 2022, 9, 2105373. [Google Scholar] [CrossRef] [PubMed]
  75. Garner, J.; Skidmore, S.; Park, H.; Park, K.; Choi, S.; Wang, Y. A protocol for assay of poly (lactide-co-glycolide) in clinical products. Int. J. Pharm. 2015, 495, 87–92. [Google Scholar] [CrossRef]
  76. Wang, R.; Bao, Q.; Clark, A.G.; Wang, Y.; Zhang, S.; Burgess, D.J. Characterization and in vitro release of minocycline hydrochloride microspheres prepared via coacervation. Int. J. Pharm. 2022, 628, 122292. [Google Scholar] [CrossRef] [PubMed]
  77. Huang, C.L.; Kumar, S.; Tan, J.J.; Boey, F.Y.; Venkatraman, S.S.; Steele, T.W.; Loo, J.S. Modulating drug release from poly (lactic-co-glycolic acid) thin films through terminal end-groups and molecular weight. Polym. Degrad. Stab. 2013, 98, 619–626. [Google Scholar] [CrossRef]
  78. Lanao, R.P.F.; Jonker, A.M.; Wolke, J.G.; Jansen, J.A.; van Hest, J.C.; Leeuwenburgh, S.C. Physicochemical properties and applications of poly (lactic-co-glycolic acid) for use in bone regeneration. Tissue Eng. Part B Rev. 2013, 19, 380–390. [Google Scholar] [CrossRef]
  79. Zare, E.N.; Jamaledin, R.; Naserzadeh, P.; Afjeh-Dana, E.; Ashtari, B.; Hosseinzadeh, M.; Vecchione, R.; Wu, A.; Tay, F.R.; Borzacchiello, A.; et al. Metal-based nanostructures/PLGA nanocomposites: Antimicrobial activity, cytotoxicity, and their biomedical applications. ACS Appl. Mater. Interfaces 2019, 12, 3279–3300. [Google Scholar] [CrossRef]
  80. Locatelli, E.; Comes Franchini, M. Biodegradable PLGA-b-PEG polymeric nanoparticles: Synthesis, properties, and nanomedical applications as drug delivery system. J. Nanopart. Res. 2012, 14, 1316. [Google Scholar] [CrossRef]
  81. Yu, B.; Pu, Y.; Liu, J.; Liao, J.; Chen, K.; Zhang, J.; Zhong, W.; Hu, Y.; Wang, X.Q.; Liu, B.; et al. Targeted delivery of emodin to adipocytes by aptamer-functionalized PEG-PLGA nanoparticles in vitro. J. Drug Deliv. Sci. Technol. 2020, 57, 101739. [Google Scholar] [CrossRef]
  82. Sheffey, V.V.; Siew, E.B.; Tanner, E.E.; Eniola-Adefeso, O. PLGA’s plight and the role of stealth surface modification strategies in its use for intravenous particulate drug delivery. Adv. Healthc. Mater. 2022, 11, 2101536. [Google Scholar] [CrossRef]
  83. Ramalho, M.J.; Serra, É.; Lima, J.; Loureiro, J.A.; Pereira, M.C. Chitosan-PLGA mucoadhesive nanoparticles for gemcitabine repurposing for glioblastoma therapy. Eur. J. Pharm. Biopharm. 2024, 200, 114326. [Google Scholar] [CrossRef]
  84. Zhou, J.; Yi, T.; Zhang, Z.; Yu, D.G.; Liu, P.; Wang, L.; Zhu, Y. Electrospun Janus core (ethyl cellulose//polyethylene oxide)@ shell (hydroxypropyl methyl cellulose acetate succinate) hybrids for an enhanced colon-targeted prolonged drug absorbance. Adv. Compos. Hybrid Mater. 2023, 6, 189. [Google Scholar] [CrossRef]
  85. Soares, D.C.F.; Domingues, S.C.; Viana, D.B.; Tebaldi, M.L. Polymer-hybrid nanoparticles: Current advances in biomedical applications. Biomed. Pharmacother. 2020, 131, 110695. [Google Scholar]
  86. Malcor, J.D.; Mallein-Gerin, F. Biomaterial functionalization with triple-helical peptides for tissue engineering. Acta Biomater. 2022, 148, 1–21. [Google Scholar] [CrossRef] [PubMed]
  87. Chen, Y.; Zhou, W.; Xia, Y.; Zhang, W.; Zhao, Q.; Li, X.; Gao, H.; Liang, Z.; Ma, G.; Yang, K.; et al. Targeted cross-linker delivery for the in situ mapping of protein conformations and interactions in mitochondria. Nat. Commun. 2023, 14, 3882. [Google Scholar] [CrossRef]
  88. Barros, C.H.; Hiebner, D.W.; Fulaz, S.; Vitale, S.; Quinn, L.; Casey, E. Synthesis and self-assembly of curcumin-modified amphiphilic polymeric micelles with antibacterial activity. J. Nanobiotechnol. 2021, 19, 104. [Google Scholar] [CrossRef]
  89. Wei, Y.; Liu, Z.; Zhu, X.; Jiang, L.; Shi, W.; Wang, Y.; Xu, N.; Gang, F.; Wang, X.; Zhao, L.; et al. Dual directions to address the problem of aseptic loosening via electrospun PLGA@ aspirin nanofiber coatings on titanium. Biomaterials 2020, 257, 120237. [Google Scholar] [CrossRef]
  90. Monteleone, P.; Cascino, G.; Rossi, A.; Rocca, P.; Bertolino, A.; Aguglia, E.; Amore, M.; Andriola, I.; Bellomo, A.; Biondi, M.; et al. Evolution of antipsychotic-induced extrapyramidal symptoms in patients with schizophrenia in the real-life: A 4-year follow-up naturalistic study. Schizophr. Res. 2022, 248, 279–286. [Google Scholar] [CrossRef] [PubMed]
  91. Wang, X.; Bao, Q.; Wang, R.; Kwok, O.; Maurus, K.; Wang, Y.; Qin, B.; Burgess, D.J. In situ forming risperidone implants: Effect of PLGA attributes on product performance. J. Control. Release 2023, 361, 777–791. [Google Scholar] [CrossRef]
  92. Shen, J.; Choi, S.; Qu, W.; Wang, Y.; Burgess, D.J. In vitro-in vivo correlation of parenteral risperidone polymeric microspheres. J. Control. Release 2015, 218, 2–12. [Google Scholar] [CrossRef]
  93. Andhariya, J.V.; Shen, J.; Choi, S.; Wang, Y.; Zou, Y.; Burgess, D.J. Development of in vitro-in vivo correlation of parenteral naltrexone loaded polymeric microspheres. J. Control. Release 2017, 255, 27–35. [Google Scholar] [CrossRef]
  94. Liu, J.C.T.; De La Peña, R.; Tocol, C.; Sattely, E.S. Reconstitution of early paclitaxel biosynthetic network. Nat. Commun. 2024, 15, 1419. [Google Scholar] [CrossRef] [PubMed]
  95. Luiz, M.T.; Abriata, J.P.; Raspantini, G.L.; Tofani, L.B.; Fumagalli, F.; de Melo, S.M.G.; da Silva Emery, F.; Swiech, K.; Marcato, P.D.; Lee, R.; et al. In vitro evaluation of folate-modified PLGA nanoparticles containing paclitaxel for ovarian cancer therapy. Mater. Sci. Eng. C 2019, 105, 110038. [Google Scholar] [CrossRef]
  96. Khaled, S.S.; Soliman, H.A.; Abdel-Gabbar, M.; Ahmed, N.A.; Attia, K.A.H.A.; Mahran, H.A.; El-Nahass, E.S.; Ahmed, O.M. The preventive effects of naringin and naringenin against paclitaxel-induced nephrotoxicity and cardiotoxicity in male Wistar rats. Evid.-Based Complement. Altern. Med. 2022, 2022, 8739815. [Google Scholar] [CrossRef]
  97. Xu, D.; Song, X.J.; Chen, X.; Wang, J.W.; Cui, Y.L. Advances and future perspectives of intranasal drug delivery: A scientometric review. J. Control. Release 2024, 367, 366–384. [Google Scholar] [CrossRef] [PubMed]
  98. Huang, M.; Liu, J.; Fan, Y.; Sun, J.; Cheng, J.X.; Zhang, X.F.; Zhai, B.T.; Guo, D.Y. Development of curcumin-loaded galactosylated chitosan-coated nanoparticles for targeted delivery of hepatocellular carcinoma. Int. J. Biol. Macromol. 2023, 253, 127219. [Google Scholar] [CrossRef]
  99. Rawal, S.; Patel, M.M. Threatening cancer with nanoparticle aided combination oncotherapy. J. Control. Release 2019, 301, 76–109. [Google Scholar] [CrossRef]
  100. Hu, H.; Liao, Z.; Xu, M.; Wan, S.; Wu, Y.; Zou, W.; Wu, J.; Fan, Q. Fabrication, optimization, and evaluation of paclitaxel and curcumin coloaded PLGA nanoparticles for improved antitumor activity. ACS Omega 2022, 8, 976–986. [Google Scholar] [CrossRef]
  101. Shi, M.; McHugh, K.J. Strategies for overcoming protein and peptide instability in biodegradable drug delivery systems. Adv. Drug Deliv. Rev. 2023, 199, 114904. [Google Scholar] [CrossRef] [PubMed]
  102. Hamadani, C.M.; Dasanayake, G.S.; Gorniak, M.E.; Pride, M.C.; Monroe, W.; Chism, C.M.; Heintz, R.; Jarrett, E.; Singh, G.; Edgecomb, S.X.; et al. Development of ionic liquid-coated PLGA nanoparticles for applications in intravenous drug delivery. Nat. Protoc. 2023, 18, 2509–2557. [Google Scholar] [CrossRef]
  103. Müller, M.; Vörös, J.; Csucs, G.; Walter, E.; Danuser, G.; Merkle, H.; Spencer, N.; Textor, M. Surface modification of PLGA microspheres. J. Biomed. Mater. Res. Part A 2003, 66, 55–61. [Google Scholar] [CrossRef]
  104. Ochi, M.; Wan, B.; Bao, Q.; Burgess, D.J. Influence of PLGA molecular weight distribution on leuprolide release from microspheres. Int. J. Pharm. 2021, 599, 120450. [Google Scholar] [CrossRef] [PubMed]
  105. Zhu, J.; Qin, F.; Ji, Z.; Fei, W.; Tan, Z.; Hu, Y.; Zheng, C. Mannose-modified PLGA nanoparticles for sustained and targeted delivery in hepatitis B virus immunoprophylaxis. AAPS PharmSciTech 2020, 21, 13. [Google Scholar] [CrossRef] [PubMed]
  106. Islan, G.A.; Rodenak-Kladniew, B.; Noacco, N.; Duran, N.; Castro, G.R. Prodigiosin: A promising biomolecule with many potential biomedical applications. Bioengineered 2022, 13, 14227–14258. [Google Scholar] [CrossRef] [PubMed]
  107. Obayemi, J.; Danyuo, Y.; Dozie-Nwachukwu, S.; Odusanya, O.; Anuku, N.; Malatesta, K.; Yu, W.; Uhrich, K.; Soboyejo, W. PLGA-based microparticles loaded with bacterial-synthesized prodigiosin for anticancer drug release: Effects of particle size on drug release kinetics and cell viability. Mater. Sci. Eng. C 2016, 66, 51–65. [Google Scholar] [CrossRef]
  108. Tian, X.; Zhu, H.; Du, S.; Zhang, X.Q.; Lin, F.; Ji, F.; Tsou, Y.H.; Li, Z.; Feng, Y.; Ticehurst, K.; et al. Injectable PLGA-coated ropivacaine produces a long-lasting analgesic effect on incisional pain and neuropathic pain. J. Pain 2021, 22, 180–195. [Google Scholar] [CrossRef]
  109. Gao, G.; Sun, X.; Liang, G. Nanoagent-promoted mild-temperature photothermal therapy for cancer treatment. Adv. Funct. Mater. 2021, 31, 2100738. [Google Scholar] [CrossRef]
  110. Shao, J.; Xie, H.; Huang, H.; Li, Z.; Sun, Z.; Xu, Y.; Xiao, Q.; Yu, X.F.; Zhao, Y.; Zhang, H.; et al. Biodegradable black phosphorus-based nanospheres for in vivo photothermal cancer therapy. Nat. Commun. 2016, 7, 12967. [Google Scholar] [CrossRef]
  111. Yan, J.; Shan, C.; Zhang, Z.; Li, F.; Sun, Y.; Wang, Q.; He, B.; Luo, K.; Chang, J.; Liang, Y. Autophagy-induced intracellular signaling fractional nano-drug system for synergistic anti-tumor therapy. J. Colloid Interface Sci. 2023, 645, 986–996. [Google Scholar] [CrossRef] [PubMed]
  112. Wang, C.; Shi, X.; Song, H.; Zhang, C.; Wang, X.; Huang, P.; Dong, A.; Zhang, Y.; Kong, D.; Wang, W. Polymer-lipid hybrid nanovesicle-enabled combination of immunogenic chemotherapy and RNAi-mediated PD-L1 knockdown elicits antitumor immunity against melanoma. Biomaterials 2021, 268, 120579. [Google Scholar] [CrossRef]
  113. Zhang, X.; Dong, Y.; Zeng, X.; Liang, X.; Li, X.; Tao, W.; Chen, H.; Jiang, Y.; Mei, L.; Feng, S.S. The effect of autophagy inhibitors on drug delivery using biodegradable polymer nanoparticles in cancer treatment. Biomaterials 2014, 35, 1932–1943. [Google Scholar] [CrossRef]
  114. Danhier, F.; Feron, O.; Préat, V. To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J. Control. Release 2010, 148, 135–146. [Google Scholar] [CrossRef] [PubMed]
  115. Yue, Z.G.; Wei, W.; Lv, P.P.; Yue, H.; Wang, L.Y.; Su, Z.G.; Ma, G.H. Surface charge affects cellular uptake and intracellular trafficking of chitosan-based nanoparticles. Biomacromolecules 2011, 12, 2440–2446. [Google Scholar] [CrossRef]
  116. Koerner, J.; Horvath, D.; Herrmann, V.L.; MacKerracher, A.; Gander, B.; Yagita, H.; Rohayem, J.; Groettrup, M. PLGA-particle vaccine carrying TLR3/RIG-I ligand Riboxxim synergizes with immune checkpoint blockade for effective anti-cancer immunotherapy. Nat. Commun. 2021, 12, 2935. [Google Scholar] [CrossRef] [PubMed]
  117. Wu, D.; Chen, Q.; Chen, X.; Han, F.; Chen, Z.; Wang, Y. The blood–brain barrier: Structure, regulation, and drug delivery. Signal Transduct. Target. Ther. 2023, 8, 217. [Google Scholar] [CrossRef]
  118. Terstappen, G.C.; Meyer, A.H.; Bell, R.D.; Zhang, W. Strategies for delivering therapeutics across the blood–brain barrier. Nat. Rev. Drug Discov. 2021, 20, 362–383. [Google Scholar] [CrossRef]
  119. Biswaro, L.S.; Garcia, M.P.; da Silva, J.R.; Neira Fuentes, L.F.; Vera, A.; Escobar, P.; Azevedo, R.B. Itraconazole encapsulated PLGA-nanoparticles covered with mannose as potential candidates against leishmaniasis. J. Biomed. Mater. Res. Part B Appl. Biomater. 2019, 107, 680–687. [Google Scholar] [CrossRef] [PubMed]
  120. Handa, M.; Sanap, S.N.; Bhatta, R.S.; Patil, G.P.; Ghose, S.; Singh, D.P.; Shukla, R. Combining donepezil and memantine via mannosylated PLGA nanoparticles for intranasal delivery: Characterization and preclinical studies. Biomater. Adv. 2023, 154, 213663. [Google Scholar] [CrossRef]
  121. Jeong, S.H.; Jang, J.H.; Lee, Y.B. Drug delivery to the brain via the nasal route of administration: Exploration of key targets and major consideration factors. J. Pharm. Investig. 2023, 53, 119–152. [Google Scholar] [CrossRef] [PubMed]
  122. Yavarpour-Bali, H.; Ghasemi-Kasman, M.; Pirzadeh, M. Curcumin-loaded nanoparticles: A novel therapeutic strategy in treatment of central nervous system disorders. Int. J. Nanomed. 2019, 14, 4449–4460. [Google Scholar] [CrossRef]
  123. Maiti, P.; Dunbar, G.L. Use of curcumin, a natural polyphenol for targeting molecular pathways in treating age-related neurodegenerative diseases. Int. J. Mol. Sci. 2018, 19, 1637. [Google Scholar] [CrossRef]
  124. Hegde, M.; Girisa, S.; BharathwajChetty, B.; Vishwa, R.; Kunnumakkara, A.B. Curcumin Formulations for Better Bioavailability: What We Learned from Clinical Trials Thus Far? ACS Omega 2023, 8, 10713–10746. [Google Scholar] [CrossRef] [PubMed]
  125. Wu, J.; Wang, J.; Zhang, J.; Zheng, Z.; Kaplan, D.L.; Li, G.; Wang, X. Oral delivery of curcumin using silk nano-and microparticles. ACS Biomater. Sci. Eng. 2018, 4, 3885–3894. [Google Scholar] [CrossRef] [PubMed]
  126. Zhang, X.; Yao, W.; Zhou, H.; Wang, H.; Kong, B.; Bai, F. Ginkgo biloba extract-loaded PLGA microcapsules generated from microfluidics for Alzheimer’s disease treatment. Mater. Des. 2024, 238, 112735. [Google Scholar] [CrossRef]
  127. Zhang, X.; Shi, Y.; Zhang, Z.; Yang, Z.; Huang, G. Intra-articular delivery of tetramethylpyrazine microspheres with enhanced articular cavity retention for treating osteoarthritis. Asian J. Pharm. Sci. 2018, 13, 229–238. [Google Scholar] [CrossRef] [PubMed]
  128. Lin, C.Y.; Hsieh, Y.T.; Chan, L.Y.; Yang, T.Y.; Maeda, T.; Chang, T.M.; Huang, H.C. Dictamnine delivered by PLGA nanocarriers ameliorated inflammation in an oxazolone-induced dermatitis mouse model. J. Control. Release 2021, 329, 731–742. [Google Scholar] [CrossRef]
  129. Karami, Z.; Mehrzad, J.; Akrami, M.; Hosseinkhani, S. Anti-inflammation-based treatment of atherosclerosis using Gliclazide-loaded biomimetic nanoghosts. Sci. Rep. 2023, 13, 13880. [Google Scholar] [CrossRef]
  130. Vargason, A.M.; Anselmo, A.C.; Mitragotri, S. The evolution of commercial drug delivery technologies. Nat. Biomed. Eng. 2021, 5, 951–967. [Google Scholar] [CrossRef]
  131. McHugh, K.J.; Guarecuco, R.; Langer, R.; Jaklenec, A. Single-injection vaccines: Progress, challenges, and opportunities. J. Control. Release 2015, 219, 596–609. [Google Scholar] [CrossRef]
  132. Watkins, H.C.; Pagan, C.L.; Childs, H.R.; Posada, S.; Chau, A.; Rios, J.; Guarino, C.; DeLisa, M.P.; Whittaker, G.R.; Putnam, D. A single dose and long lasting vaccine against pandemic influenza through the controlled release of a heterospecies tandem M2 sequence embedded within detoxified bacterial outer membrane vesicles. Vaccine 2017, 35, 5373–5380. [Google Scholar] [CrossRef]
  133. Danaeifar, M.; Negahdari, B.; Eslam, H.M.; Zare, H.; Ghanaat, M.; Koushali, S.S.; Malekshahi, Z.V. Polymeric nanoparticles for DNA vaccine-based cancer immunotherapy: A review. Biotechnol. Lett. 2023, 45, 1053–1072. [Google Scholar] [CrossRef]
  134. Rubsamen, R.; Herst, C.; Lloyd, P.; Heckerman, D. Eliciting cytotoxic T-lymphocyte responses from synthetic vectors containing one or two epitopes in a C57BL/6 mouse model using peptide-containing biodegradable microspheres and adjuvants. Vaccine 2014, 32, 4111–4116. [Google Scholar] [CrossRef] [PubMed]
  135. Rosalia, R.A.; Cruz, L.J.; van Duikeren, S.; Tromp, A.T.; Silva, A.L.; Jiskoot, W.; de Gruijl, T.; Löwik, C.; Oostendorp, J.; van der Burg, S.H.; et al. CD40-targeted dendritic cell delivery of PLGA-nanoparticle vaccines induce potent anti-tumor responses. Biomaterials 2015, 40, 88–97. [Google Scholar] [CrossRef]
  136. Silva, A.; Soema, P.; Slütter, B.; Ossendorp, F.; Jiskoot, W. PLGA particulate delivery systems for subunit vaccines: Linking particle properties to immunogenicity. Hum. Vaccines Immunother. 2016, 12, 1056–1069. [Google Scholar] [CrossRef]
  137. Tran, K.T.; Gavitt, T.D.; Farrell, N.J.; Curry, E.J.; Mara, A.B.; Patel, A.; Brown, L.; Kilpatrick, S.; Piotrowska, R.; Mishra, N.; et al. Transdermal microneedles for the programmable burst release of multiple vaccine payloads. Nat. Biomed. Eng. 2021, 5, 998–1007. [Google Scholar] [CrossRef] [PubMed]
  138. Jin, S.; Xia, X.; Huang, J.; Yuan, C.; Zuo, Y.; Li, Y.; Li, J. Recent advances in PLGA-based biomaterials for bone tissue regeneration. Acta Biomater. 2021, 127, 56–79. [Google Scholar] [CrossRef]
  139. Kim, H.J.; Han, M.A.; Shin, J.Y.; Jeon, J.H.; Lee, S.J.; Yoon, M.Y.; Kim, H.J.; Choi, E.J.; Do, S.H.; Yang, V.C.; et al. Intra-articular delivery of synovium-resident mesenchymal stem cells via BMP-7-loaded fibrous PLGA scaffolds for cartilage repair. J. Control. Release 2019, 302, 169–180. [Google Scholar] [CrossRef]
  140. Xu, T.M.; Chu, H.Y.; Li, M.; Talifu, Z.; Ke, H.; Pan, Y.Z.; Xu, X.; Wang, Y.H.; Guo, W.; Wang, C.L.; et al. Establishment of FK506-Enriched PLGA Nanomaterial Neural Conduit Produced by Electrospinning for the Repair of Long-Distance Peripheral Nerve Injury. J. Nanomater. 2022, 2022, 3530620. [Google Scholar] [CrossRef]
  141. Wang, Y.; Kankala, R.K.; Cai, Y.Y.; Tang, H.X.; Zhu, K.; Zhang, J.T.; Yang, D.Y.; Wang, S.B.; Zhang, Y.S.; Chen, A.Z. Minimally invasive co-injection of modular micro-muscular and micro-vascular tissues improves in situ skeletal muscle regeneration. Biomaterials 2021, 277, 121072. [Google Scholar] [CrossRef]
  142. Lü, J.M.; Wang, X.; Marin-Muller, C.; Wang, H.; Lin, P.H.; Yao, Q.; Chen, C. Current advances in research and clinical applications of PLGA-based nanotechnology. Expert Rev. Mol. Diagn. 2009, 9, 325–341. [Google Scholar] [CrossRef]
  143. Ramezanpour, S.; Tavatoni, P.; Akrami, M.; Navaei-Nigjeh, M.; Shiri, P. Potential wound healing of PLGA nanoparticles containing a novel L-Carnitine–GHK peptide conjugate. J. Nanomater. 2022, 2022, 6165759. [Google Scholar] [CrossRef]
  144. Blanco-Fernandez, B.; Castano, O.; Mateos-Timoneda, M.Á.; Engel, E.; Pérez-Amodio, S. Nanotechnology approaches in chronic wound healing. Adv. Wound Care 2021, 10, 234–256. [Google Scholar] [CrossRef] [PubMed]
  145. Stromberg, Z.R.; Jacobsen, D.E.; Kocheril, P.A.; Kubicek-Sutherland, J.Z. Chapter 17 - Biological toxicity and environmental hazards associated with PLGA nanoparticles. In Poly(lactic-co-glycolic acid) (PLGA) Nanoparticles for Drug Delivery; Kesharwani, P., Ed.; Micro and Nano Technologies; Elsevier: Amsterdam, The Netherlands, 2023; pp. 457–475. [Google Scholar] [CrossRef]
  146. Yoo, J.; Won, Y.Y. Phenomenology of the initial burst release of drugs from PLGA microparticles. ACS Biomater. Sci. Eng. 2020, 6, 6053–6062. [Google Scholar] [CrossRef] [PubMed]
  147. Hamdallah, S.I.; Zoqlam, R.; Yang, B.; Campbell, A.; Booth, R.; Booth, J.; Belton, P.; Qi, S. Using a systematic and quantitative approach to generate new insights into drug loading of PLGA nanoparticles using nanoprecipitation. Nanoscale Adv. 2024, 6, 3188–3198. [Google Scholar] [CrossRef] [PubMed]
  148. Yang, F.; Wang, W.; Zhou, J.; Yu, Z.; An, M.; He, W.; Xue, Y.; Chen, F. Transdermal delivery of IBU-PLGA nanoparticles with dissolving microneedle array patch. J. Drug Deliv. Sci. Technol. 2024, 95, 105528. [Google Scholar] [CrossRef]
  149. Bausart, M.; Rodella, G.; Dumont, M.; Ucakar, B.; Vanvarenberg, K.; Malfanti, A.; Préat, V. Combination of local immunogenic cell death-inducing chemotherapy and DNA vaccine increases the survival of glioblastoma-bearing mice. Nanomed. Nanotechnol. Biol. Med. 2023, 50, 102681. [Google Scholar] [CrossRef]
  150. Casanova, E.A.; Rodriguez-Palomo, A.; Stähli, L.; Arnke, K.; Gröninger, O.; Generali, M.; Neldner, Y.; Tiziani, S.; Dominguez, A.P.; Guizar-Sicairos, M.; et al. SAXS imaging reveals optimized osseointegration properties of bioengineered oriented 3D-PLGA/aCaP scaffolds in a critical size bone defect model. Biomaterials 2023, 294, 121989. [Google Scholar] [CrossRef]
  151. Huang, Y.; Ye, K.; He, A.; Wan, S.; Wu, M.; Hu, D.; Xu, K.; Wei, P.; Yin, J. Dual-layer conduit containing VEGF-A–Transfected Schwann cells promotes peripheral nerve regeneration via angiogenesis. Acta Biomater. 2024, 180, 323–336. [Google Scholar] [CrossRef]
Figure 2. Impact of the PLGA L: G ratio on solubility. PLGAs with varying L: G ratios breaking down in various solvents [57]. (A) Characterization of PLGA involves assessing its molecular weight, L: G ratio, and shape, which can be linear or branched. When a formulation contains a mixture of PLGAs with varying properties (as shown in the dotted circle), determining and characterizing each PLGA component becomes difficult. (B) PLGAs with varying L:G ratios can dissolve in different solvents, enabling the separation of PLGAs even when their molecular weights are identical. (C) The serum leuprolide concentrations in human patients after injection of Eligard, which provides 45 mg of leuprolide for a 6-month effective treatment. (D) Assessment of the branch unit numbers in Glu-PLGAs and standard star-PLGAs with 3, 4, 5, and 6 arms. All Glu-PLGAs maintain a 55:45 L: G ratio. Copyright (2019), Elsevier.
Figure 2. Impact of the PLGA L: G ratio on solubility. PLGAs with varying L: G ratios breaking down in various solvents [57]. (A) Characterization of PLGA involves assessing its molecular weight, L: G ratio, and shape, which can be linear or branched. When a formulation contains a mixture of PLGAs with varying properties (as shown in the dotted circle), determining and characterizing each PLGA component becomes difficult. (B) PLGAs with varying L:G ratios can dissolve in different solvents, enabling the separation of PLGAs even when their molecular weights are identical. (C) The serum leuprolide concentrations in human patients after injection of Eligard, which provides 45 mg of leuprolide for a 6-month effective treatment. (D) Assessment of the branch unit numbers in Glu-PLGAs and standard star-PLGAs with 3, 4, 5, and 6 arms. All Glu-PLGAs maintain a 55:45 L: G ratio. Copyright (2019), Elsevier.
Polymers 16 02606 g002
Figure 3. Promising vaccine delivery vehicle: mannose-modified PLGA nanoparticle (MNP)-loaded hepatitis B surface antigen (HBsAg) protein highly presents to lymphocytes, which target and induce long-lasting immunity. (A) Schematic representation of the in vivo fate of nanoparticles. (B) Flow cytometry histogram of in vitro cellular uptake in HaCaT, BMDCs, and RAW 264.7 cells. Data are presented as mean ± SD (n = 3). * p < 0.05. (C) Splenocyte proliferation after in vitro co-incubation with Ag [105]. Copyright (2019), Springer Nature.
Figure 3. Promising vaccine delivery vehicle: mannose-modified PLGA nanoparticle (MNP)-loaded hepatitis B surface antigen (HBsAg) protein highly presents to lymphocytes, which target and induce long-lasting immunity. (A) Schematic representation of the in vivo fate of nanoparticles. (B) Flow cytometry histogram of in vitro cellular uptake in HaCaT, BMDCs, and RAW 264.7 cells. Data are presented as mean ± SD (n = 3). * p < 0.05. (C) Splenocyte proliferation after in vitro co-incubation with Ag [105]. Copyright (2019), Springer Nature.
Polymers 16 02606 g003
Figure 4. An example of PLGA application for pain treatment. (A) Schematic drawing of in vivo experiments on Sprague–Dawley (SD) rats with chronic constriction injury (CCI). PLGA-CD-DEX-RVC nanofibers were implanted into the sciatic nerve and measured by an acetone test to confirm the cold allodynia response. The mitigating mechanism of neuropathic pain is blocking of the transient receptor potential vanilloid 1 (TRPV1) channel and inhibition of microglia. (B) Luminescence imaging after transplantation of the PLGA and PLGA-CD-DEX-RVC nanofibers. (C) Immunofluorescence staining of TRPV1 and NeuN in DRG. Scale bar: 20 μm [8]. Copyright (2022), Elsevier.
Figure 4. An example of PLGA application for pain treatment. (A) Schematic drawing of in vivo experiments on Sprague–Dawley (SD) rats with chronic constriction injury (CCI). PLGA-CD-DEX-RVC nanofibers were implanted into the sciatic nerve and measured by an acetone test to confirm the cold allodynia response. The mitigating mechanism of neuropathic pain is blocking of the transient receptor potential vanilloid 1 (TRPV1) channel and inhibition of microglia. (B) Luminescence imaging after transplantation of the PLGA and PLGA-CD-DEX-RVC nanofibers. (C) Immunofluorescence staining of TRPV1 and NeuN in DRG. Scale bar: 20 μm [8]. Copyright (2022), Elsevier.
Polymers 16 02606 g004
Figure 5. An example of PLGA application for cancer treatment. (A) Schematic illustration of the mechanism whereby PLGA–biotin conjugate irinotecan nanoparticles target colon cancer. (B) In vitro cellular uptake study followed by confocal analysis. (C) Corrected total cell fluorescence of Cy5.5 quantified from images using NIH ImageJ software (ImageJ Freeware; https://imagej.net/ij/, 8 October 2023) and represented in a bar graph (mean ± SEM). (D) Near-infrared fluorescence (NIRF) images of the tumors and major organs dissected from PLGA-NP-DiR and PLGA-B-NP-DiR-treated mice. Source: [17].
Figure 5. An example of PLGA application for cancer treatment. (A) Schematic illustration of the mechanism whereby PLGA–biotin conjugate irinotecan nanoparticles target colon cancer. (B) In vitro cellular uptake study followed by confocal analysis. (C) Corrected total cell fluorescence of Cy5.5 quantified from images using NIH ImageJ software (ImageJ Freeware; https://imagej.net/ij/, 8 October 2023) and represented in a bar graph (mean ± SEM). (D) Near-infrared fluorescence (NIRF) images of the tumors and major organs dissected from PLGA-NP-DiR and PLGA-B-NP-DiR-treated mice. Source: [17].
Polymers 16 02606 g005
Figure 8. Examples of PLGA applications for vaccine development. (A) Schematic illustrations of fully implanted core-shell microneedles (MNs) that deliver their payloads in pulses to mimic drug release from multiple bolus injections over a long period. (B) Optical IVIS analysis images demonstrating the pulsatile release of the core-shell microneedles embedded inside the skin of rats over a long period. (C) Optical images of the skin of rats from groups that received Prevnar-13 core-shell MNs, s.c. injections, and blank MNs. (D) The immune response of rats that received the Prevnar-13-loaded core-shell MNs, multiple bolus s.c. injections, and blank MNs. Data are presented as mean ± SEM; n = 5 rats per group; independent experiments. Statistical analysis was performed using a two-way ANOVA with repeated measures; *** p < 0.001; NS, adjusted p = 0.7973 [137]. Copyright (2020), Springer Nature.
Figure 8. Examples of PLGA applications for vaccine development. (A) Schematic illustrations of fully implanted core-shell microneedles (MNs) that deliver their payloads in pulses to mimic drug release from multiple bolus injections over a long period. (B) Optical IVIS analysis images demonstrating the pulsatile release of the core-shell microneedles embedded inside the skin of rats over a long period. (C) Optical images of the skin of rats from groups that received Prevnar-13 core-shell MNs, s.c. injections, and blank MNs. (D) The immune response of rats that received the Prevnar-13-loaded core-shell MNs, multiple bolus s.c. injections, and blank MNs. Data are presented as mean ± SEM; n = 5 rats per group; independent experiments. Statistical analysis was performed using a two-way ANOVA with repeated measures; *** p < 0.001; NS, adjusted p = 0.7973 [137]. Copyright (2020), Springer Nature.
Polymers 16 02606 g008
Figure 9. Examples of PLGA applications for tissue regeneration. (A) Schematic illustration of the construction of C2C12-laden PLGA PMs by dynamic culture method. (B) Evaluation of the in vivo myogenic performance of minimally invasively injected composite microtissues in severe combined immunodeficiency (SCID) mice. (C) CLSM images showing the formation of C2C12 aggregation with prolonged culture time (7 d). (D) CCK-8 assay assessing the numbers of HUVECs encapsulated in the vessel-like PEG HMs. **** p < 0.0001. (E) CLSM images showing the longitudinal cross-section of PEG HMs without cells [141]. Copyright (2021), Elsevier.
Figure 9. Examples of PLGA applications for tissue regeneration. (A) Schematic illustration of the construction of C2C12-laden PLGA PMs by dynamic culture method. (B) Evaluation of the in vivo myogenic performance of minimally invasively injected composite microtissues in severe combined immunodeficiency (SCID) mice. (C) CLSM images showing the formation of C2C12 aggregation with prolonged culture time (7 d). (D) CCK-8 assay assessing the numbers of HUVECs encapsulated in the vessel-like PEG HMs. **** p < 0.0001. (E) CLSM images showing the longitudinal cross-section of PEG HMs without cells [141]. Copyright (2021), Elsevier.
Polymers 16 02606 g009
Table 1. List of commercial PLGA drug delivery products approved by the FDA.
Table 1. List of commercial PLGA drug delivery products approved by the FDA.
Product name,
Manufacturer
APITypeApprobsl Date(s), Indication(s)Dose
Vivitrol®
Alkermes, Inc., Dublin, Ireland
NaltrexoneMicroparticleAlcohol dependence,
relapse in opioid dependence
380 mg per month
Zoladex® Depot
AstraZeneca, Cambridge, UK
Goserelin acetateImplantBreast cancer, prostate cancer, endometriosis3.6 mg/10.8 per month
Lupron Depot®
AbbVie Inc., North Chicago, IL, USA
Leuprolide acetateMicrosphereEndometriosis, advanced prostate cancer7.5 mg per month
Lupron®
AbbVie Inc., North Chicago, IL, USA
Leuprolide acetateMicrosphereEndometriosis3.75 mg per month
Eligard®
Tolmar, Inc., Fort Collins, CO, USA
Leuprolide acetateIn situ gelProstate cancer symptoms7.5 mg a month
22.5 mg per 3 months
30 mg for 4 months
45 mg per 6 months
Sandostatin® LAR
Novartis, Basel, Switzerland
Octreotide acetateMicrosphereAcromegaly, flushing episodes and watery diarrhea
(caused by vasoactive intestinal peptide tumors)
10/20/30 mg per month
Atridox®
Atrix, Inc., Fort Collins, CO, USA
Doxycycline hyclateIn situ gelChronic periodontitis.42.5 mg per week
Nutropin Depot®
Genentech, Inc., South San Francisco, CA, USA
SomatotropinMicroparticleGrowth failure, growth hormone deficiency13.5/18/22.5 mg per month
Trelstar®
Tolmar, Inc., Fort Collins, CO, USA
Triptorelin pamoateMicroparticleAdvanced prostate cancer relief treatment3.75 mg a month
11.25 mg per 3 months
22.5 mg for 6 months
Somatuline® Depot
Ipsen, Inc., Basking Ridge, NJ, USA
LanreotideMicroparticleAcromegaly,
symptoms caused by neuroendocrine tumors
60 mg per month
Arestin®
OraPharma, Inc., Warminster, PA, USA
Minocycline HClMicroparticlePeriodontitis1 mg per 2 weeks
Risperidal® Consta
Janssen, Inc., Titusville, NJ, USA
RisperidoneMicroparticleSchizophrenia12.5/25/37.5/50 mg per 2 weeks
Perseris™
Indivior Inc., Richmond, VA, USA
RisperidoneIn situ gelSchizophrenia90/120 mg per month
Ozurdex®
Allergan, Dublin, Ireland
DexamethasoneImplantMacular edema, diabetic macular edema,
non-infectious uveitis
0.7 mg variable dosing frequency
Propel®
Intersect ENT, Inc., Menlo Park, CA, USA
Mometasone furoateImplantChronic sinusitis0.37 mg per month
Bydureon®
AstraZeneca, Cambridge, UK
ExenatideMicroparticleType 2 diabetes mellitus2.0 mg per week
Signifor® LAR
Novartis, Basel, Switzerland
PasireotideMicroparticleAlcohol dependence,
relapse in opioid dependence
380 mg per month
Zilretta®
Flexion, Inc., Burlington, MA, USA
Triamcinolone acetoamideMicroparticleOsteoarthritis32 mg per 3 months
Sublocade™
Indivior Inc., Berkshire, UK
Buprenorphinein situ gelModerate-to-severe opioid use disorder100/300 mg per month
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

Yang, J.; Zeng, H.; Luo, Y.; Chen, Y.; Wang, M.; Wu, C.; Hu, P. Recent Applications of PLGA in Drug Delivery Systems. Polymers 2024, 16, 2606. https://doi.org/10.3390/polym16182606

AMA Style

Yang J, Zeng H, Luo Y, Chen Y, Wang M, Wu C, Hu P. Recent Applications of PLGA in Drug Delivery Systems. Polymers. 2024; 16(18):2606. https://doi.org/10.3390/polym16182606

Chicago/Turabian Style

Yang, Jie, Huiying Zeng, Yusheng Luo, Ying Chen, Miao Wang, Chuanbin Wu, and Ping Hu. 2024. "Recent Applications of PLGA in Drug Delivery Systems" Polymers 16, no. 18: 2606. https://doi.org/10.3390/polym16182606

APA Style

Yang, J., Zeng, H., Luo, Y., Chen, Y., Wang, M., Wu, C., & Hu, P. (2024). Recent Applications of PLGA in Drug Delivery Systems. Polymers, 16(18), 2606. https://doi.org/10.3390/polym16182606

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