Next Article in Journal
The Evolution of Single-Cell RNA Sequencing Technology and Application: Progress and Perspectives
Next Article in Special Issue
Biodegradation Studies of Polyhydroxybutyrate and Polyhydroxybutyrate-co-Polyhydroxyvalerate Films in Soil
Previous Article in Journal
The Relationship between Serum miRNAs and Early Mortality in Multiple Myeloma Patients Treated with Bortezomib-Based Regimens
Previous Article in Special Issue
The Influence of Constraints on Gelation in a Controlling/Living Copolymerization Process
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Overview of Antimicrobial Biodegradable Polyester-Based Formulations

by
Oana Gherasim
1,
Valentina Grumezescu
1,2 and
Stefan Andrei Irimiciuc
1,2,*
1
National Institute for Lasers, Plasma and Radiation Physics, 077125 Magurele, Romania
2
Academy of Romanian Scientists, Splaiul Independentei No. 54, 050044 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2945; https://doi.org/10.3390/ijms24032945
Submission received: 29 November 2022 / Revised: 18 January 2023 / Accepted: 21 January 2023 / Published: 2 February 2023
(This article belongs to the Collection Frontiers in Polymeric Materials)

Abstract

:
As the clinical complications induced by microbial infections are known to have life-threatening side effects, conventional anti-infective therapy is necessary, but not sufficient to overcome these issues. Some of their limitations are connected to drug-related inefficiency or resistance and pathogen-related adaptive modifications. Therefore, there is an urgent need for advanced antimicrobials and antimicrobial devices. A challenging, yet successful route has been the development of new biostatic or biocide agents and biomaterials by considering the indisputable advantages of biopolymers. Polymers are attractive materials due to their physical and chemical properties, such as compositional and structural versatility, tunable reactivity, solubility and degradability, and mechanical and chemical tunability, together with their intrinsic biocompatibility and bioactivity, thus enabling the fabrication of effective pharmacologically active antimicrobial formulations. Besides representing protective or potentiating carriers for conventional drugs, biopolymers possess an impressive ability for conjugation or functionalization. These aspects are key for avoiding malicious side effects or providing targeted and triggered drug delivery (specific and selective cellular targeting), and generally to define their pharmacological efficacy. Moreover, biopolymers can be processed in different forms (particles, fibers, films, membranes, or scaffolds), which prove excellent candidates for modern anti-infective applications. This review contains an overview of antimicrobial polyester-based formulations, centered around the effect of the dimensionality over the properties of the material and the effect of the production route or post-processing actions.

1. Introduction

With the aim to overcome the current challenges of classical pharmacotherapy (drug-related pharmacological mechanisms, such as partial specificity for receptor targeting, limited control, and distribution over tissue volume, but also inappropriate or incorrect administration) [1,2,3], emerging bio-nanotechnologies enable the progress of patient-oriented and performance-enhanced therapeutic strategies. Modern pharmacotherapy relies on the synergetic association between biomedical sciences (biochemistry and biophysics, cellular and molecular biology, and physiological and pathological molecular interactions) and nanotechnology (in-depth understanding and accurate manipulation of ultra-small-scaled mater and materials), towards developing the necessary infrastructure for the implementation of personalized healthcare desideratum [4,5,6].
The progress of alternative antimicrobial therapies, which involve interdisciplinary research, implies challenging and prospective protocols that study microbial interactions [7,8,9], investigate biocidal and biostatic mechanisms [10,11,12], and develop functional anti-pathogenic treatments [13,14,15]. The use of biopolymers as unconventional antimicrobial platforms (either as intrinsic anti-pathogenic agents [16,17,18] or as active drug carriers [19,20,21]) is of great significance when considering conventional antibiotherapy efficacy control, combating the alarming occurrence of drug-resistant pathogens, and limiting or eliminating the clinical implications of biofilm-related complications.
When developing new and effective antimicrobials, polymeric platforms provide indisputable advantages regarding the manufacturing of modern pharmacologically active formulations. The main targeted aspects are the protection of embedded drugs, targeted and triggered control over localized tissue distribution, specific and selective targeting of cellular receptors, reduced or mitigated collateral/side effects, and pharmacological efficacy [22,23]. In particular, the design and fabrication of pharmacological formulations based on biodegradable polyesters is of great importance for the progress of personalized biomedicine. Their intrinsic peculiarities are enabling modern and effective strategies for molecular diagnosis and treatment [24,25], anti-infective therapy [26] and cancer management [27,28], and tissue engineering and regenerative medicine [29,30].
An updated literature survey on the most recent reports in new and efficient antimicrobial formulations based on biodegradable polyesters is herein proposed. Our study considers the most explored and prolific polyester candidates for developing modern anti-infective platforms, evidencing their use as particulate, layered, or complex formulations (Figure 1).

2. Biopolyesters

The use of biodegradable polyesters in biomedicine relies on their versatile characteristics, including biomimicking composition and microstructure, tunable degradability and physiological metabolization, and complex functionality and biological performance [31,32].
The degradation of polyesters (Figure 2) represents the essential feature of such biopolymers, conferring excellent biocompatibility of polyester-based platforms (due to the physiological processing and natural elimination of the hydrolysis-resulted degradation products) and resulting in promising pharmacologically active formulations [33,34].
The degradation of biopolyesters is mediated by the (auto)catalytic hydrolytic or enzymatic degradation of constituent ester linkages and depends on specific intrinsic physicochemical aspects, such as composition and molecular weight, hydrophobicity, crystallinity, and glass transition temperature [35,36,37]. Slower drug release and delayed polymer degradation have been evidenced in the case of biopolyesters with high molecular weight (due to low chain mobility and reduced swelling and solubility) and high hydrophobicity (due to increased hydrolytic resistance) [38,39,40]. Even with the high drug-loading efficiency reported for highly crystalline polyesters, the consequences of increased crystallinity and reduced glass transition temperature (through the drug-plasticizing effect) are seen in the reduced drug release and polymer degradation rates [41,42,43].
Polylactide (PLA) and poly(lactide-co-glycolide) copolymer (PLGA) are key representatives of polyester-based therapeutics. Their intrinsic mechanical behavior, thermoplastic properties, tunable solubility and degradation, and excellent biocompatibility enable their implementation in the fabrication of modern biomedical platforms, such as biodegradable and bioresorbable implantable devices [44,45], tailorable stimuli-responsive pharmaceuticals [46,47], and patient-oriented tissue substitutes or augmentations [48,49]. In addition, retard formulations [50] and long-term medical devices [51,52] can be developed by exploring the intrinsic features of polycaprolactone (PCL) and poly(3-hydroxybutyrate-co-3-hydroxyvalerate) copolymer (PHBV), namely, thermoplasticity- and crystallinity-related hydrophobicity and the reduced degradation rate. Moreover, the piezoelectric effect of biodegradable materials and devices based on naturally derived PLA and PHBV represents a strong argument for their incorporation in smart therapeutic platforms [53,54].
The degradation of PLGA and PHBV copolymers can be tailored through the molecular weight and ratio of constituent monomers. For instance, the fastest degradation rate of PLGA copolymers is for the 50:50 lactide–to–glycolide (LA/GA) representative, which decreases with the increasing of the lactide constituent, due to the LA-mediated abundance of hydrophobic methyl side groups and GA-mediated reduced crystallinity [55,56]. In PHBV, higher degradation rates have been evidenced for copolymers with more hydroxyvalerate (HV) content, because of the HV-mediated reduced crystallinity and accelerated hydrolysis [57,58].
Additives (drugs, biomolecules, and inorganic and organic reinforcers) impact the stability and degradability of biopolyesters at a microstructural level [59,60,61], but their degradation profile can also be modulated by external factors, such as pH [62], temperature [63,64], electromagnetic radiation [65,66], and enzymes [67,68].
In [69], the effect of femtosecond laser irradiation on the biodegradability of PLGA films was shown. Comparisons of the 800 nm and 400 nm heat-affected zones for irradiation have shown that infrared irradiation provides a small affected area. The results were also corelated with the degradation rate of the PLGD when 400 nm irradiated films had a considerably higher degradation rate when compared with the 800 nm ones. The behavior was explained by the decrease in molecular weight as a result of the dissociation of the chemical bonds. The aforementioned results are amongst the first reports on the dependence of the degradation rate of biodegradable polymers on the irradiation wavelength.

3. Particulate Formulations

Biodegradable polyesters represent attractive candidates for the fabrication of new and efficient antimicrobials. Conventional and modified emulsification protocols are usually employed for the synthesis of biopolyester-based micro-/nano-sized particles [70,71], but superior outcomes have also been reported by using nano-precipitation [72,73] and microfluidic and membrane emulsion [74,75]. The proper synthesis method (the selection of which is strongly influenced by the stability, half-lifetime, hydrophilicity, or lipophilicity of the antimicrobial payload) [76,77] facilitates the formation of compact or porous spheres and capsules with intrinsic microstructures that are directly related to the degradation and release profiles.
The chemical reactivity and compositional versatility of such systems enable impressive possibilities for conjugation or functionalization (micro-/nano-particles), loading or entrapment (compact or porous micro-/nano-spheres), and encapsulation (micro-/nano-capsules) of the antimicrobial cargo, which may consist of drugs, phytochemicals, biomolecules, inorganic nanosystems, and macromolecules [78,79,80]. The same features open the path for additional surface modifications and coatings, with the final goal to fabricate multifunctional formulations with active targeting abilities. The surface of biopolyester-based particulate formulations can be (bio)chemically tailored to target microbial pathogens (electrostatic interactions or specific binding to surface molecules overexpressed by microbial cells) [81,82,83] or infected tissues (selective coupling to specific tissue receptors, including the stealth effect) [84,85,86]. Besides such targeting abilities, most polyester-based formulations exert their therapeutic action through passive targeting, which is mainly a concentration-dependent effect that occurs at the infection site due to the increased retention and accumulation caused by the vascular and lymphatic impairment [87,88,89].
PLA-based pharmacological platforms possess reduced immunogenicity (owing to their intrinsic degradation mechanisms, which results in non-toxic and metabolically active secondary products) and enable controlled and/or targeted delivery mechanisms [90,91]. As a result, several PLA-based formulations have been approved by regulatory institutions for safe use in clinical practice [92,93].
Prominent inhibitory effects on Gram-negative strains were reported by encapsulating eugenol and linalool in PLA particles [94], due to hydrophobicity-mediated interactions between particles and microbial outer membranes. By contrast, the embedding of carvacrol (53.9%) within polyethyleneimine-coated PLA nanoparticles (114.7 ± 1.02 nm) resulted in long-term antimicrobial activity against Gram-positive strains [95], through enhanced cellular uptake facilitated by the cationic-charged nanoparticles. Further, a prolonged anti-staphylococcal efficacy has been shown for gentamicin-loaded PLA microspheres [96].
In addition, in the case of bone fixing screws modified with layered coatings of PLA films and vancomycin-loaded PLA nanospheres, the long-term release of the antibiotic (up to 24 days) led to the eradication of Staphylococcus aureus (S. aureus) during the contamination phase, followed by a drastic inhibition of the staphylococcal bacterial biofilm [97].
Micro-/nano-sized PLGA-based platforms have an essential role in emerging modern pharmacotherapy, with several clinically approved formulations [98,99]. The anti-infective clinical potential of PLGA systems, evidenced by efficient drug entrapment and effective drug release, has been evidenced for various antibiotics, including ceftiofur [100], doxycycline [101,102], gentamycin [103], rapamycin [104], rifapentine [105,106], and vancomycin [107].
The efficient loading of tobramycin/dioctyl sulfosuccinate conjugates within PLGA nanoparticles (>89%) determined significant bactericidal effects against Pseudomonas aeruginosa (P. aeruginosa), even when using sub-inhibitory antibiotic concentrations [108]. Complementary studies evidenced that the antimicrobial efficacy of ciprofloxacin, florfenicol, and cefpodoxime proxetil was enhanced following their incorporation into particles of PLGA conjugated with polysorbate surfactant [109], Eudragit (enteric methacrylic polymer) [110], and (CS) chitosan/Eudragit [111], respectively.
While exhibiting important antibacterial and hemostatic effects, the sustained release (up to 2 weeks) of tylotoin peptide molecules from CS-coated PLGA nanocapsules determined reduced inflammatory events and accelerated healing in full-thickness skin wounds [112]. PLGA nanospheres were recently proposed to overcome solubility limitations of plant-derived gentiopicroside, demonstrating important anti-staphylococcal effects and accelerated healing rates in diabetic wounds [113]. Considerable bactericidal and bacteriostatic effects have also been reported by encapsulating cinnamaldehyde (33.20 ± 0.85%) in PLGA/CS nanoparticles [114], while porous PLGA microspheres embedding antimicrobial peptides showed promising potential for the local management of bone infections [115].
Composite PLGA/CS microspheres provided a prolonged release of antimicrobial peptides and determined subsequent long-lasting antibacterial effects (almost 3 months) against microorganisms from the oral flora [116]. Aiming to reduce the cariogenic risk, effective anti-streptococcal platforms have been developed by incorporating chlorhexidine salts into PLGA microparticles [117] and PLGA-coated mesoporous silica nanoparticles [118]. The prophylactic potential of PLGA nanoparticles modified with antimicrobial peptides on periodontal disease has also been highlighted; such platforms prevented the adhesion of oral microorganisms to the endogenous streptococcal community and, consequently, inhibited the formation and development of polymicrobial biofilms [119].
Bacteriophage-loaded PLGA microparticles exhibited pronounced bactericidal effects against planktonic and sessile P. aeruginosa. Following their murine inhalation, the as-developed systems induced a drastic reduction of the pulmonary microbial community and an effective control over pneumonia-associated pulmonary and hepatic complications [120]. The intracellular release of clarithromycin [121] and amikacin-moxifloxacin complex [122] from PLGA nanocapsules has shown promising potential to combat opportunistic infections associated with lung disease. Inhalable platforms based on curcumin-loaded PLGA nanoparticles embedded in drug matrix (tobramycin/ciprofloxacin/azithromycin and N-acetylcysteine) have been proposed for the multivalent treatment of lung infections, as evidenced by their cumulative anti-inflammatory, antibacterial, and mucolytic effects [123].
The triggered release of clarithromycin from hybrid microparticles based on the magnesium core, antibiotic-loaded PLGA layer, and CS coating significantly reduced the gastric level of Helicobacter pylori [124]. With a similar goal, the targeted therapeutic potential of amoxicillin-loaded nanocapsules based on PLGA and CS derivatives has been reported [125]. PLGA nanocapsules loaded with meropenem–cyclodextrin complex provided antibiotic stability under acidic conditions and enabled controlled antibiotic release under neutral conditions. As a result, they have been proposed as efficient anti-infective platforms for the gastrointestinal tract [126]. Considerable anti-amoebic effects have been reported for the efficient encapsulation of gallic acid (82.86%) into PLGA particles (~100 nm) [127].
Though it is more difficult to safely assess their efficiency, promising antiviral outcomes of biopolyester-based formulations have also been reported (Table 1).
Highly stable PCL-based micelles showed efficient loading of luteolin (97.3% ± 1.1%) and ofloxacin (64.23%) and determined their prolonged release (up to 8 hours), being proposed as tablet formulations for the local treatment of fungal infections [143] or as particulate systems for treating ocular infections [144], respectively. Sustained and pH-responsive drug release was evidenced for negatively charged PCL nanoparticles encapsulating cefotaxime, determining important anti-fouling activity against bacterial pathogens [145].
The surface coating of urinary catheters with chlorhexidine-loaded PCL nanospheres (152 ± 37 nm) [146] and PEG-block-PCL micelles (40.21 ± 3.85 nm) [147] proved an effective and prolonged strategy for reducing the contamination by and colonization of uropathogenic microorganisms. Complementary studies evidenced that nanosystems-based multilayer coatings determined long-term antibacterial and anti-biofilm effects through the sustained release of chlorhexidine (for up to 4 weeks), while exhibiting good biocompatibility and reducing the longevity-related limitations of catheterization (encrustation and crystal deposition) [26,148].
The particular degradation kinetics of PHBV-based biomaterials are beneficial for the successful development of therapeutically effective formulations, with promising outcomes for tissue engineering [52,149] and modern pharmacotherapy [150,151].
Impressive therapeutic efficacy and preventive action have been reported in Salmonella Typhimurium systemic infection following the intramuscular administration of ceftiofur-loaded PHBV microparticles. The pharmacokinetic and toxicological studies revealed no changes in the biochemical and hematological parameters, and a lack of hepatotoxic and nephrotoxic effects, respectively [152]. Nano-magnetite-loaded PHBV/ceftiofur composite nanoparticles significantly inhibited the development of Escherichia coli (E. coli), while exhibiting high cytocompatibility in human hepatocytes. The as-developed hybrid nanosystems (243.0 ± 17 nm) have been evaluated as multifunctional platforms for the local management of infections, by means of magnetic resonance imaging, magnetic hyperthermia, and controlled release of the antibiotic [153]. The multi-faceted functionality of PHBV–Fe3O4 (magnetite) composites have also been reported in the case of biopolymer microspheres loaded with magnetic nanoparticles functionalized with lauric and oleic acids [154,155].
The encapsulation of epirubicin within composite PHBV–PEG (polyethylene glycol) particles determined important antibacterial effects against Gram-positive and Gram-negative strains, with superior efficiency to equivalent concentrations of free drug. The obtained nanosystems (152.3 ± 0.6 nm) exhibited fast and sustained pH-dependent drug release, as evidenced under acidic and neutral physiologically simulated conditions (2 and 8 days, respectively) [156]. Highly cytocompatible PHBV–CS spheres proved to have potentiating effects on different bioproduced antibiotics (against various clinically relevant bacterial strains); however, only kanamycin-loaded composites exhibited reduced pro-inflammatory effects beneficial for the modulation of the healing process and microbicidal mechanisms.
Owing to their superior mechanical properties and tunable degradability, PHBV-based formulations are extensively investigated regarding the development of biomaterials and devices for restorative and regenerative applications of bone tissue. With the aim to reduce the bacterial contamination and colonization susceptibility of metallic implants, levofloxacin-loaded PHBV microspheres were embedded within alginate matrix and validated as compact coatings that exert sustained bactericidal effects against E. coli [157]. Superior antimicrobial efficacy and prolonged release of cinnamaldehyde (7 days) and vancomycin (4 days) have been reported in the case of PHBV-based microspheres embedded with mesoporous vitroceramic nanoparticles [158] or loaded within vitroceramic scaffolds [159], respectively.
The effects of WS2 nanotubes addition of the mechanical properties of biodegradable polymers (PLLA) were also investigated [160]. The best improvement of the mechanical properties are shown for INT–WS2 addition up to 0.5 wt. %. The addition of nanotubes in the composition of polymers reportedly also reduces the friction coefficient of the polymer–nanotube composite.
Additionally, no hindering of viscosity or polymer matrix moduli is reported based on rheological sties performed on the composites. The polymers’ bond stretching was highlighted by the use of Raman Spectroscopy, with no observable interference with respect to the polymerization process by the insertion of WS2 nanotubes. Increases in the PLLA’s crystallinity are also reported by use of differential scanning calorimetry investigations and confirmed by X-ray diffraction with nanotubes acting as nucleation centers, thus transforming the composite into a semi-crystalline material.
More than extending the safe use of conventional antimicrobials and limiting their negative side effects, biopolyester-based particle formulations represent ideal candidates for modern anti-infective therapy, contributing to the emerging clinical evaluation of more effective, comfortable, and compliant treatments. Intrinsic and circumstantial biodegradability, but also excellent biocompatibility and thermoplasticity, represent key aspects that highlight their promising use for the development of modern and efficient antimicrobial platforms, susceptible to various administration routes. Being biosafe and biodegradable materials, polyesters have a great potential for the commercial fabrication of particulate antimicrobials, providing specific, selective, controlled, targeted, and personalized anti-infective effects.

4. Layered Formulations

Fabricating protective and highly biocompatible surface-modifying coatings [161] and nano-sized/-structured bidimensional formulations [162,163] is an attractive and emerging strategy to modulate the microbial susceptibility of commercial medical devices and develop new anti-infective devices, respectively. Given their tunable biomechanics, thermophysics, and biochemistry, but also their versatile processability, polyesters are indisputable candidates for the fabrication of such active carriers or enhancers for local antimicrobial effects.
Various synthesis methods have been employed to obtain antimicrobial coatings for biomedical materials and devices [164,165] (Figure 3). The degradation and release profiles of such formulations can be tuned at a microstructural and morphological level, depending on the therapeutic effect and final use. Furthermore, boosted bioactivity and additional functionality may be achieved by means of polyester coatings.
PLA films were reported to have the role of active matrices for the release of metallic (silver) and oxide (zinc and titanium oxide) nanoparticles with intrinsic anti-pathogenic activity, exhibiting pronounced inhibitory effects against the E. coli strain [166]. By contrast, increased anti-staphylococcal efficacy was obtained in the case of layered films of PLA and thymol-encapsulated zein-chitosan solid particles [167]. Enhanced anti-biofilm activity against S. aureus was also demonstrated for PLA films embedded with stearate-stabilized silver nanoparticles [168] and nano-magnetite conjugated with eucalyptus essential oil [169], while exhibiting excellent biocompatibility with respect to human-derived endothelial cells and mesenchymal stem cells, respectively.
Excellent bioactivity and prolonged anti-staphylococcal efficacy have been reported for hybrid structures based on PLA films reinforced with gentamicin-oaded coralline hydroxyapatite (HAp) nanoparticles (as evidenced up to 4 weeks [170,171]) and PLA–PVA (polyvinyl alcohol) microsphere coatings entrapping usnic acid [172].
The immobilization of recombinant antimicrobial peptides in PLA membranes has been successfully evaluated for topical bactericidal use, with additional beneficial effects on the adhesion and proliferation of human fibroblasts [173]. Concerning the fabrication of antimicrobial wound dressings, PLA/gelatin nanofiber mats demonstrated a sustained release of phyto-conjugated silver nanoparticles, but also proper mechanical and gelation properties [174]. Antibacterial and antioxidant activity have been also reported for PLA–PEG composite films incorporating silver nanoparticles conjugated with phytochemicals [175], while PLA/PEG nanofibrous mats were recently proposed for the transdermal administration of acyclovir against HSV type-1 infection [176]. Non-toxic nanocomposites with ultraviolet-light barrier properties have been developed by the impregnation of cinnamaldehyde within PLA/lignin nanoparticle films [177].
An increased antimicrobial efficiency has also been reported in the case of biodegradable PLGA membranes loaded with phytochemicals, such as cinnamaldehyde and carvacrol [178], aloe vera [179], and thymol [180]. The immobilization of eugenol and clove essential oil within PLGA films was beneficial for potentiated anti-biofilm activity against enterohemorrhagic E. coli [181]. Excellent efficiency against mature microbial biofilms has been evidenced in the case of nanostructured coatings based on PLGA microspheres or PLGA films embedded with phytochemical-conjugated [182] and antibiotic-functionalized magnetite nanoparticles [183,184]. Superior anti-staphylococcal efficacy was also reported for hybrid coatings of PLGA–PVA microspheres loaded with usnic acid and nano-magnetite [185].
Using graphene oxide filler proved effective for increasing the hydrophilicity and modulating the adsorption ability of biomolecules (antimicrobial peptides and growth factors) in PLGA films. Such nanostructured formulations showed important antibacterial effects against opportunistic strains and accelerated healing and tissue regeneration ability, being evaluated as promising platforms for the infection-free regeneration of wounds [186]. Superior mechanical properties and enhanced acyclovir loading efficiency were reported for PLGA/PCL nanofibrous mats, in comparison with bare PLGA materials. Besides exhibiting short-term viral inhibition, the reduced polymer degradation and continuous drug release resulted in long-term protection against viral transmission of HSV type-2 infection [187].
HAp/PLGA coatings entrapping ceftriaxone and cefuroxime are suitable materials for the normal development of osteoblasts [188] and showed enhanced anti-biofilm activity against the E. coli strain. Bacterial inhibition has also been demonstrated in the case of multi-layered membranes consisting of either collagen nanofibers loaded with PLGA nanoparticles and aspirin or curcumin-functionalized collagen nanofibers. Besides their antibacterial efficiency, the as-developed hybrid structures exhibited important osteogenic activity, being proposed for reducing infection-associated complications in guided bone regeneration [189]. Hybrid films consisting of vancomycin-loaded chitosan hydrogel and PLGA nanoparticles loaded with osteoinductive protein enabled the sequential release of biosubstances (2 and 12 days, respectively) and resulted in accelerated and uncomplicated osteointegration [190]. Therapeutic effects (stable prosthetic fixation and faster healing) and prophylactic effects (effective control of methicillin-resistant S. aureus contamination for up to 28 days) were revealed by the encapsulation of linezolid within PLGA nanofibrous membranes [191].
PCL films loaded with ciclopirox olamine-modified vermiculite determined the long-term inhibition of bacterial and fungal biofilms, while the addition of zinc oxide (ZnO) nanoparticles resulted in potentiated anti-biofilm effects [192]. Highly hemocompatible PCL films embedded with copper oxide nanoparticles have been evaluated as promising candidates for the management of diabetic foot ulcers infected with opportunistic methicillin-resistant S. aureus [193].
PCL/poly(ethylene succinate) polyester mixture provided extended release of the biocide agent and prolonged antibacterial effects when used as a shell coating for drug-loaded polyvinylpyrrolidone core nanofibers, leading to the development of bacteria-degradable nanofibrous membranes for wound care management [194]. As hydrophilicity and water retention are important aspects when designing wound dressings, the addition of gelatin proved beneficial for increasing these parameters in PCL nanofibers. Meshes based on ciprofloxacin-loaded PCL core/tetracycline-loaded gelatin shell fibers showed improved mechanical properties (when compared to uniaxial membranes) and exhibited long-lasting antibacterial effects [195]. Also aiming at the development of efficient topical platforms, biocompatible PCL nanofiber arrays decorated with ZnO tetrapod nanoparticles demonstrated important and sustained dose-dependent antiviral effects against both type-1 and type-2 HSV [196]. The successful revaluation of anti-pathogenic phytochemicals in the development of antimicrobial alternatives has been demonstrated in the case of PHBV films loaded with rosemary and green tea extracts [197], oregano essential oil [198,199], and eugenol-encapsulated mesoporous silica nanoparticles [200].
Nanostructured coatings of PHBV–PVA microspheres entrapping eugenol-functionalized nano-magnetite exhibited important anti-adherence and sustained anti-biofilm effects (evidenced against S. aureus and P. aeruginosa), showing superior cytocompatibility with respect to human endothelial cells [201]. Prolonged anti-biofilm efficiency against Gram-positive and Gram-negative pathogens, as well as excellent biological behavior on osteoblasts and endothelial cells, was reported for coatings based on lysozyme-embedded PHBV/PEG microspeheres [202].
With the aim to fabricate performance-enhanced topical formulations, PHBV nanofibrous membranes were reinforced with hybrid systems consisting of cellulose nanocrystals (NCC) and ZnO, which resulted in improved mechanical strength and thermal stability, and antibacterial efficiency (demonstrated against both E. coli and S. aureus strains) [203]. In a similar study, the cumulative release of tetracycline (~80%) from PHBV and NCC composite membranes grafted with methacrylic polymer was attained after 4 days of testing under physiologically simulated conditions. Under weakly acidic conditions or by increasing the temperature, the antibiotic release period was reduced to only 2 hours [204]. The reinforcement of PHBV/alginate films with graphene nanoplatelets led to the formation of thermally stable, highly hydrophobic, and electrically conductive biomaterials, which exhibited important action against a bacteriophage-based model of enveloped viruses [205].
Volova et al. [206] investigated the effect of CO2 laser irradiation of PHA’s films produced by the solvent cast technique. Two different working modes were considered—a continuous wave using a power of 3 W and a scanning speed of 2 m/s and a pulsed wave using a power of 13.5 W and a scanning speed of 1 m/s—for the irradiation of poly-3-hydroxybutyrate in a mixture with 30% 4-hydroxybutyrate, 3-hydroxyvalerate, or 3-hydroxyhexanoate. The polyhydroxyalkanoates (PHA) films offer a wide range of thermal, mechanical, or molecular properties, with the irradiation affecting all of their key parameters on top of biocompatibility. For example, the poly-3-hydroxybutyrate (P(3HB)) films present a decrease in contact angle from 92 to 80, while both the surface energy and the roughness increase from 30 to 57 mN/m and from 144 up to a maximum value of 290 nm, respectively. All PHA’s films irradiated in continuous mode present a decrease in contact angle down to 80 and an overall roughness increase up to 45 mN/m. The pulsed irradiation regime defines stronger morphological changes, as expected due to the higher local beam power, and thus a steeper decrease in contact angle down to 67. Assessing the cell metabolic activity for a culture of mouse fibroblast proved the advantage of pulsed treatment, which increased the number of viable cells with a factor of 1.5. These new results offer the perspective of targeted surface modification for cell attachment control.

5. Complex Formulations

The successful use of biopolyesters in fabricating more complex constructs (scaffolds, sponges, and foams) and topical formulations (gels) has also been evidenced [207,208].
Owing to their facile processability and impressive clinical outcomes, biodegradable polyesters have been extensively used for fabricating three-dimensional constructs for implantable devices and regenerative medicine. More than representing adequate mechanical and biochemical support for beneficial interactions with physiological biomolecules and resident cells, such constructs own the indisputable structural advantage. The porous microstructure of scaffold-type formulations facilitates local nutrient transport and provides adequate biomimetic support for cellular ingrowth, but also promotes local vascularization and tissue repair/regeneration.
Highly biocompatible and bioactive systems able to recover the integrity and functionality of damaged tissues through their restoration, replacement, or regeneration can be fabricated by properly adjusting the composition, microstructure, wettability, surface charge, morphology, topography, and reactivity of biopolyester-based platforms [209,210]. In addition, their advanced biofunctionality can be modulated by using composite constructs of polyesters and natural polymers, such as polysaccharides and proteins [211,212].
Excellent bioactivity and prolonged anti-staphylococcal efficacy have been reported for hybrid structures based on collagen, nano-Hap, and vancomycin-loaded PLA scaffolds (18 days) [213]. Highly biocompatible architectures based on PLA, barium sulphate particles embedded in polydopamine, and levofloxacin with excellent inhibitory activity against the development of S. aureus have been proposed as bone fixation devices [214].
The incorporation of green indocyanine into PLA nanofibrous networks has been attempted in order to eradicate bacterial contamination and colonization of chronic wounds by means of photodynamic therapy. The newly developed materials are exhibiting accelerated degradation under alkaline conditions or in the presence of proteases, promoting physiological cellular events and encouraging pro-angiogenic effects [215].
Electrospun PLGA scaffolds modified with growth factor and antimicrobial peptide have been proposed for the accelerated healing of cutaneous wounds, while reducing the risk for opportunistic contamination with E. coli and S. aureus [216]. Considerable antimicrobial effects have been reported by the immobilization of ciprofloxacin within PLGA/alginate nanofibrous networks [217] and PLGA nanoparticles embedded into PVA hydrogels [218]. Moreover, demineralized bone matrix loaded with PLGA microparticles co-encapsulating vancomycin and HAp nanoparticles was proposed as osteogenic and highly efficient antibacterial fillers for infected bone defects [219].
For altering the hydrophobic nature of polyester, PCL/gelatin [220,221,222] and PCL/chitosan [223] composite scaffolds, with suitable mechanical behavior for skin tissue use, were developed. The porosity-related features (release profile, swelling, and permeability) of such constructs were beneficial for infection-free wound-healing applications when loaded with antibiotics and phytochemicals. Similar outcomes were also evidenced for juglone-modified PCL scaffolds [224]. Improved biomechanics and hydrophilicity were reported for PCL/gelatin scaffolds reinforced with calcium phosphate-modified graphene oxide. Exhibiting important antibacterial activity, the clindamycin-loaded osteoinductive scaffolds represent promising candidates as electrically actuated bone substitutes [225].
The release of vancomycin over a period of 22 days was evidenced in the case of composite structures based on PHBV, nano-diamond, and nano-HAp [226]. Recently, bilayer PHBV/pullulan nanofibrous scaffolds were developed as bacteria-repellent formulations for wound-healing applications. While the polyester layer provided suitable microstructure for increased water and oxygen retention and suitable architecture for cellular proliferation and migration, the hydrophilic polysaccharide layer acted as a protective membrane against bacterial transmission [227]. Enhanced and sustained antibacterial activity has been evidenced by cephalexin-loaded PHBV nanofibrous sheets against different methicillin-resistant S. aureus strains, both in cellular and animal models. The as-developed dressing material has been evaluated as a biosafe platform for the efficient treatment of opportunistic infections in chronic diabetic foot ulcers [228].
Besides representing active carriers or enhancers for local antimicrobial treatment, The biomechanical compliance, biomimetic microstructure, and bioresorbable ability of such formulations represent key aspects promoting biodegradable polyester in designing and fabricating functional tissue substitutes.
Though exhibiting optimal biomechanical properties and biodegradability, the hydrophobicity, slow degradation rate, and drug release profiles of biopolyester-based formulations should be properly tuned for specific biomedical uses. In this regard, their modification with highly hydrophilic and stimuli-responsive polymers represents an attractive strategy to fabricate advanced platforms, as confirmed by several in vivo and ex vivo studies (Table 2).
Magnetic nanoparticles have been at the center of an impressive number of studies, as they have been categorized as reliable candidates for a wide range of applications in biomedicine. Their attractive intrinsic physical properties, often in combination with desired high biocompatibility and low immunogenicity, have the potential to tackle modern issues in nanotechnology and materials engineering directed towards biomedicine. Recent reports [240,241,242] have presented different routes to improve and highlight the properties of these magnetic nanoparticles, either by thin film coatings, sensors, or drug carriers or as components in polymer structures. Various applications have been developed based on their unique properties in order to induce spatial displacement on nanometer scales for cell seeding, materials engineering, or targeted drug delivery [243]. While there are reports [244,245,246,247] that present control over the functionality of certain cells by using magnetic nanoparticles, limitations have risen from adverse effects on the cells, which are yet to be understood [248]. Recent reports [249,250,251] on magnetic actuation have shown that there are promising results towards controlling mesenchymal stromal cells or other cells aimed at bone or cardiovascular issues, for example, bone tissue engineering performed by means of magnetic nanocomposite (nanoparticle-embedded polymers) engineering via thin films, scaffolds, or implants [252]. The main goal of these studies is to obtain differential selectivity for specific molecules towards cell functionalization control under magnetic field action [253,254,255].
Simultaneously, a continuous effort has been performed towards regenerative medicine by controlling the healing and repair process for certain biological structures by phenotypic modulation of stromal cells via matrix tailoring of the physical properties or by molecular targeting of intercellular paths [256,257,258,259,260,261,262]. The development of materials with smart dynamics having the ability to aid regenerative processes at a cellular level is also done by external reactive processes, such as optical or thermoelectric stimulation, or by self-regeneration and enzymatic sensitivity control [263,264,265,266,267]. While the literature concerning tissue healing by magnetic field manipulation at a cellular level is generous, the results mostly relate to specific conditions and are very often experiment dependent in terms of the type of cell used or the stimulation of the cell with matrices with magnetic response [268,269,270]. The advantages of using magnetic nanocomposite for cell modulation have been the main driving forces behind state-of-the-art research concerning innovating and developing new strategies for tissue repair. Most utilized routes involve the control of cell behavior by functionalization, surface modulation, and cell-environment tailoring. In [271], the manufacturing of hydrogels responsive to anisotropic magnetic field tissue engineering is reported, indicating a strong correlation between the physical properties of the polymer matrix and those of the applied magnetic field. Further development of nanoparticle–polymer composites was focused mainly on increasing the response and sensitivity to the magnetic field, together with lowering the chances of biological tissue poisoning by controlling the number of magnetic nanoparticles and the cytotoxicity.
The production of spherical Pd nanoparticles generated by pulsed laser ablation in liquid (PLAL) is reported in [272]. Different routes for NP control in size and crystallinity are attempted by utilizing different solvents, as well as different wavelengths for irradiation. Multivariable control over NP production led to the generation of NP with an average dimension of 6 nm. When the antimicrobial activity was investigated, it was reported that NP produced with 1024 nm and in methanol had a better response to Staphylococcus aureus. The antimicrobial activity is understood as Pd ions release from the NP coating, with no reported harmful effects to the cells. Besides the elevated antimicrobial activity, the encouraging report of cytocompatibility shown by estimating the bactericidal factor promotes further biological testing for these promising nanostructures.

6. Conclusions

To overcome the clinical complications induced by microbial infections, known to have life-threatening side effects, conventional anti-infective therapy is generally preferred. Yet, one should note its important shortcomings concerning drug-related inefficiency or resistance and pathogen-related adaptive modifications. In this respect, advanced antimicrobials and antimicrobial devices are urgently needed.
Besides their role as protective or potentiating carriers for conventional drugs, biopolymers are characterized by an impressive ability for conjugation or functionalization, which proves beneficial to avoid collateral and side effects, and to provide targeted and triggered drug delivery, specific and selective cellular targeting, and pharmacological efficacy. It should be mentioned that biopolymers can be fabricated in various forms, i.e., particles, fibers, thin films, membranes, or scaffolds, which are demonstrated to be excellent candidates for modern anti-infective applications.
This is a comprehensive study that gathers the recent antimicrobial, polyester-based formulations, centered around the effect of the dimensionality, production route, or post processing actions on the properties of the investigated materials.

Author Contributions

Conceptualization, V.G.; writing—original draft preparation, O.G., V.G. and S.A.I.; writing—review and editing, V.G. and S.A.I.; visualization, O.G., V.G. and S.A.I.; project administration, V.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hossen, S.; Hossain, M.K.; Basher, M.; Mia, M.; Rahman, M.; Uddin, M.J. Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity studies: A review. J. Adv. Res. 2019, 15, 1–18. [Google Scholar] [CrossRef]
  2. Tran, T.T.; Tran, P.H. Nanoconjugation and encapsulation strategies for improving drug delivery and therapeutic efficacy of poorly water-soluble drugs. Pharmaceutics 2019, 11, 325. [Google Scholar] [CrossRef] [PubMed]
  3. Gherasim, O.; Popescu, R.C.; Gherasim, T.G.; Grumezescu, V.; Andronescu, E. Pharmacotherapy and nanotechnology. In Nanoparticles in Pharmacotherapy; Grumezescu, A.M., Ed.; Elsevier: Oxford, UK, 2019; pp. 1–21. [Google Scholar]
  4. Huang, H.; Feng, W.; Chen, Y.; Shi, J. Inorganic nanoparticles in clinical trials and translations. Nano Today 2020, 35, 100972. [Google Scholar] [CrossRef]
  5. Spirescu, V.A.; Chircov, C.; Grumezescu, A.M.; Andronescu, E. Polymeric nanoparticles for antimicrobial therapies: An up-to-date overview. Polymers 2021, 13, 724. [Google Scholar] [CrossRef] [PubMed]
  6. Xie, Y.; Ma, C.; Yang, X.; Wang, J.; Long, G.; Zhou, J. Phytonanomaterials as therapeutic agents and drug delivery carriers. Adv. Drug Deliv. Rev. 2021, 176, 113868. [Google Scholar] [CrossRef] [PubMed]
  7. Moghimi, M.; Bakhtiari, R.; Mehrabadi, J.F.; Jamshidi, N.; Jamshidi, N.; Siyadatpanah, A.; Mitsuwan, W.; Nissapatorn, V. Interaction of human oral cancer and the expression of virulence genes of dental pathogenic bacteria. Microb. Pathog. 2020, 149, 104464. [Google Scholar] [CrossRef]
  8. Oglou, E.O.; Karaca, B.; Kibar, H.; Haliscelik, O.; Kiran, F. The role of microbiota-derived postbiotic mediators on biofilm formation and quorum sensing-mediated virulence of Streptococcus mutans: A perspective on preventing dental caries. Microb. Pathog. 2022, 164, 105390. [Google Scholar] [CrossRef]
  9. Mashamba, T.G.; Adeosun, I.J.; Baloyi, I.T.; Tshikalange, E.T.; Cosa, S. Quorum sensing modulation and inhibition in biofilm forming foot ulcer pathogens by selected medicinal plants. Heliyon 2022, 8, e09303. [Google Scholar] [CrossRef]
  10. Vaidya, M.Y.; McBain, A.J.; Butler, J.A.; Banks, C.E.; Whitehead, K.A. Antimicrobial Efficacy and Synergy of Metal Ions against Enterococcus faecium, Klebsiella pneumoniae and Acinetobacter baumannii in Planktonic and Biofilm Phenotypes. Sci. Rep. 2017, 7, 5911. [Google Scholar] [CrossRef]
  11. Opoku-Temeng, C.; Naclerio, G.A.; Mohammad, H.; Dayal, N.; Abutaleb, N.S.; Seleem, M.N.; Sintim, H.O. N-(1,3,4-oxadiazol-2-yl)benzamide analogs, bacteriostatic agents against methicillin- and vancomycin-resistant bacteria. Eur. J. Med. Chem. 2018, 155, 797–805. [Google Scholar] [CrossRef]
  12. Biswas, D.; Tiwari, M.; Tiwari, V. Molecular mechanism of antimicrobial activity of chlorhexidine against carbapenem-resistant Acinetobacter baumannii. PLoS ONE 2019, 14, e0224107. [Google Scholar] [CrossRef] [PubMed]
  13. Buwalda, S.; Rotman, S.; Eglin, D.; Moriarty, F.; Bethry, A.; Garric, X.; Guillaume, O.; Nottelet, B. Synergistic anti-fouling and bactericidal poly(ether ether ketone) surfaces via a one-step photomodification. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 111, 110811. [Google Scholar] [CrossRef] [PubMed]
  14. Tarawneh, O.; Mahfouz, H.A.; Hamadneh, L.; Deeb, A.A.; Al-Sheikh, I.; Alwahsh, W.; Abed, A.F. Assessment of persistent antimicrobial and anti-biofilm activity of p-HEMA hydrogel loaded with rifampicin and cefixime. Sci. Rep. 2022, 12, 3900. [Google Scholar] [CrossRef] [PubMed]
  15. Zou, Y.; Chen, X.; Sun, Y.; Li, P.; Xu, M.; Fang, P.; Zhang, S.; Yuan, G.; Deng, X.; Hu, H. Antibiotics-free nanoparticles eradicate Helicobacter pylori biofilms and intracellular bacteria. J. Control. Release 2022, 348, 370–385. [Google Scholar] [CrossRef]
  16. Romanò, C.L.; De Vecchi, E.; Bortolin, M.; Morelli, I.; Drago, L. Hyaluronic Acid and Its Composites as a Local Antimicrobial/Antiadhesive Barrier. J. Bone Jt. Infect. 2017, 2, 63–72. [Google Scholar] [CrossRef]
  17. Makhayeva, D.; Filippov, S.K.; Yestemes, S.S.; Irmukhametova, G.S.; Khutoryanskiy, V.V. Polymeric iodophors with poly(2-ethyl-2-oxazoline) and poly(N-vinylpyrrolidone): Optical, hydrodynamic, thermodynamic, and antimicrobial properties. Eur. Polym. J. 2022, 165, 111005. [Google Scholar] [CrossRef]
  18. Li, X.; İlk, S.; Liu, Y.; Raina, D.B.; Demircan, D.; Zhang, B. Nonionic nontoxic antimicrobial polymers: Indole-grafted poly(vinyl alcohol) with pendant alkyl or ether groups. Polym. Chem. 2022, 13, 2307–2319. [Google Scholar] [CrossRef]
  19. Lei, D.; Wang, Q.; Kong, Y.; Chen, Y.; Luo, X. Triclosan-loaded pH-responsive copolymer to target bacteria and to have long bacteriostatic efficacy. Eur. J. Pharm. Sci. 2020, 148, 105320. [Google Scholar] [CrossRef]
  20. Yates-Alston, S.; Sarkar, S.; Cochran, M.; Kuthirummal, N.; Levi, N. Hybrid donor-acceptor polymer nanoparticles and combination antibiotic for mitigation of pathogenic bacteria and biofilms. J. Microbiol. Methods 2021, 190, 106328. [Google Scholar] [CrossRef]
  21. Pan, L.; Jiang, D.; Pan, L.; Meng, Z.; Zhuang, Y.; Huang, Y.; Ye, F.; Shi, C.; Chen, J.; Pan, J. ICAM-1-targeted and antibacterial peptide modified polymeric nanoparticles for specific combating sepsis. Mater. Des. 2022, 222, 111007. [Google Scholar] [CrossRef]
  22. Zhong, H.; Chan, G.; Hu, Y.; Hu, H.; Ouyang, D. A Comprehensive Map of FDA-Approved Pharmaceutical Products. Pharmaceutics 2018, 10, 263. [Google Scholar] [CrossRef] [PubMed]
  23. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124. [Google Scholar] [CrossRef] [PubMed]
  24. Liang, C.; Li, N.; Cai, Z.; Liang, R.; Zheng, X.; Deng, L.; Feng, L.; Guo, R.; Wei, B. Co-encapsulation of magnetic Fe3O4 nanoparticles and doxorubicin into biocompatible PLGA-PEG nanocarriers for early detection and treatment of tumours. Artif. Cells Nanomed. Biotechnol. 2019, 47, 4211–4221. [Google Scholar] [CrossRef] [PubMed]
  25. Lex, T.R.; Brummel, B.R.; Attia, M.F.; Giambalvo, L.N.; Lee, K.G.; Van Horn, B.A.; Whitehead, D.C.; Alexis, F. Iodinated Polyesters with Enhanced X-ray Contrast Properties for Biomedical Imaging. Sci. Rep. 2020, 10, 1508. [Google Scholar] [CrossRef]
  26. Srisang, S.; Wongsuwan, N.; Boongird, A.; Ungsurungsie, M.; Wanasawas, P.; Nasongkla, N. Multilayer nanocoating of Foley urinary catheter by chlorhexidine-loaded nanoparticles for prolonged release and anti-infection of urinary tract. Int. J. Polym. Mater. Polym. Biomater. 2020, 69, 1081–1089. [Google Scholar] [CrossRef]
  27. Cai, Y.; Xu, Z.; Shuai, Q.; Zhu, F.; Xu, J.; Gao, X.; Sun, X. Tumor-targeting peptide functionalized PEG-PLA micelles for efficient drug delivery. Biomater. Sci. 2020, 8, 2274–2282. [Google Scholar] [CrossRef]
  28. Zekun, P.; Jiayan, Z.; Chunyang, S. Ditelluride-Bridged PEG-PCL Copolymer as Folic Acid-Targeted and Redox-Responsive Nanoparticles for Enhanced Cancer Therapy. Front. Chem. 2020, 8, 156. [Google Scholar] [CrossRef]
  29. Wei, L.; Cai, C.; Lin, J.; Chen, T. Dual-drug delivery system based on hydrogel/micelle composites. Biomaterials 2009, 30, 2606–2613. [Google Scholar] [CrossRef]
  30. Ebrahimi, Z.; Irani, S.; Ardeshirylajimi, A.; Seyedjafari, E. Enhanced osteogenic differentiation of stem cells by 3D printed PCL scaffolds coated with collagen and hydroxyapatite. Sci. Rep. 2022, 12, 12359. [Google Scholar] [CrossRef]
  31. Washington, K.E.; Kularatne, R.N.; Karmegam, V.; Biewer, M.C.; Stefan, M.C. Recent advances in aliphatic polyesters for drug delivery applications. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2017, 9, e1446. [Google Scholar] [CrossRef]
  32. Urbánek, T.; Jäger, E.; Jäger, A.; Hrubý, M. Selectively Biodegradable Polyesters: Nature-Inspired Construction Materials for Future Biomedical Applications. Polymers 2019, 11, 1061. [Google Scholar] [CrossRef] [PubMed]
  33. Ginjupalli, K.; Shavi, G.V.; Averineni, R.K.; Bhat, M.; Udupa, N.; Nagaraja, U.P. Poly(α-hydroxy acid) based polymers: A review on material and degradation aspects. Polym. Degrad. Stab. 2017, 144, 520–535. [Google Scholar] [CrossRef]
  34. Kreua-ongarjnukool, N.; Soomherun, N.; Niyomthai, S.Y.; Chumnanvej, S. Aliphatic Polyester Nanoparticles for Drug Delivery Systems. In Smart Drug Delivery; Ahmad, U., Haider, F., Akhtar, J., Eds.; InTech: Rikeja, Croatia, 2021. [Google Scholar]
  35. Elmowafy, E.M.; Tiboni, M.; Soliman, M.E. Biocompatibility, biodegradation and biomedical applications of poly(lactic acid)/poly(lactic-co-glycolic acid) micro and nanoparticles. J. Pharm. Investig. 2019, 49, 347–380. [Google Scholar] [CrossRef]
  36. Tebaldi, M.L.; Chaves Maia, A.L.; Poletto, F.; de Andrade, F.V.; Ferreira Soares, D.C. Poly(-3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV): Current advances in synthesis methodologies, antitumor applications and biocompatibility. J. Drug Deliv. Sci. Technol. 2019, 51, 115–126. [Google Scholar] [CrossRef]
  37. Espinoza, S.M.; Patil, H.I.; Martinez, E.S.M.; Casañas Pimente, R.; Ige, P.P. Poly-ε-caprolactone (PCL), a promising polymer for pharmaceutical and biomedical applications: Focus on nanomedicine in cancer. Int. J. Polym. Mater. Polym. Biomater. 2020, 69, 85–126. [Google Scholar] [CrossRef]
  38. Liu, R.; Wang, Y.; Ma, Y.; Wu, Y.; Guo, Y.; Xu, L. Effects of the molecular weight of PLGA on degradation and drug release in vitro from an mPEG-PLGA nanocarrier. Chem. Res. Chin. Univ. 2016, 32, 848–853. [Google Scholar] [CrossRef]
  39. Kumskova, N.; Ermolenko, Y.; Osipova, N.; Semyonkin, A.; Kildeeva, N.; Gorshkova, M.; Kovalskii, A.; Kovshova, T.; Tarasov, V.; Kreuter, J.; et al. How subtle differences in polymer molecular weight affect doxorubicin-loaded PLGA nanoparticles degradation and drug release. J. Microencapsul. 2020, 37, 283–295. [Google Scholar] [CrossRef] [PubMed]
  40. Jesus, S.; Bernardi, N.; da Silva, J.; Colaço, M.; Panão Costa, J.; Fonte, P.; Borges, O. Unravelling the Immunotoxicity of Polycaprolactone Nanoparticles-Effects of Polymer Molecular Weight, Hydrolysis, and Blends. Chem. Res. Toxicol. 2020, 33, 2819–2833. [Google Scholar] [CrossRef]
  41. Vollrath, A.; Kretzer, C.; Beringer-Siemers, B.; Shkodra, B.; Czaplewska, J.A.; Bandelli, D.; Stumpf, S.; Hoeppener, S.; Weber, C.; Werz, O.; et al. Effect of Crystallinity on the Properties of Polycaprolactone Nanoparticles Containing the Dual FLAP/mPEGS-1 Inhibitor BRP-187. Polymers 2021, 13, 2557. [Google Scholar] [CrossRef]
  42. Park, K.; Otte, A.; Sharifi, F.; Garner, J.; Skidmore, S.; Park, H.; Jhon, Y.K.; Qin, B.; Wang, Y. Potential roles of the glass transition temperature of PLGA microparticles in drug release kinetics. Mol. Pharm. 2021, 18, 18–32. [Google Scholar] [CrossRef]
  43. Bikiaris, R.; Christodoulou, E.; Kostoglou, M.; Kasimatis, M.; Iatrou, H.; Nikolaidis, N. Paliperidone palmitate depot microspheres based on biocompatible poly(alkylene succinate) polyesters as long-acting injectable formulations. J. Drug Deliv. Sci. Technol. 2022, 68, 103056. [Google Scholar] [CrossRef]
  44. Menown, I.B.A.; Mamas, M.A.; Cotton, J.M.; Hildick-Smith, D.; Eberli, F.R.; Leibundgut, G.; Tresukosol, D.; Macaya, C.; Copt, S.; Sadozai Slama, S.; et al. First clinical evidence characterizing safety and efficacy of the new CoCr Biolimus-A9 eluting stent: The Biomatrix Alpha™ registry. Int. J. Cardiol. Heart Vasc. 2020, 26, 100472. [Google Scholar] [CrossRef] [PubMed]
  45. Eawsakul, K.; Tancharoen, S.; Nasongkla, N. Combination of dip coating of BMP-2 and spray coating of PLGA on dental implants for osseointegration. J. Drug Deliv. Sci. Technol. 2021, 61, 102296. [Google Scholar] [CrossRef]
  46. Dwivedi, A.; Mazumder, A.; Nasongkla, N. Layer-by-layer nanocoating of antibacterial niosome on orthopedic implant. Int. J. Pharm. 2018, 547, 235–243. [Google Scholar] [CrossRef]
  47. Zhou, J.; Hirota, K.; Ackermann, R.; Walker, J.; Wang, Y.; Choi, S.; Schwendeman, A.; Schwendeman, S.P. Reverse Engineering the 1-Month Lupron Depot®. AAPS J. 2018, 20, 105. [Google Scholar] [CrossRef]
  48. Leventis, M.D.; Fairbairn, P.; Kakar, A.; Leventis, A.D.; Margaritis, V.; Lückerath, W.; Horowitz, R.A.; Rao, B.H.; Lindner, A.; Nagursky, H. Minimally Invasive Alveolar Ridge Preservation Utilizing an In Situ Hardening β-Tricalcium Phosphate Bone Substitute: A Multicenter Case Series. Int. J. Dent. 2016, 2016, 5406736. [Google Scholar] [CrossRef]
  49. Patel, S.K.; Greene, A.C.; Desai, S.M.; Rothstein, S.; Basha, I.T.; MacPherson, J.S.; Wang, Y.; Zou, Y.; Shehabeldin, M.; Sfeir, C.S.; et al. Biorelevant and screening dissolution methods for minocycline hydrochloride microspheres intended for periodontal administration. Int. J. Pharm. 2021, 596, 120261. [Google Scholar] [CrossRef]
  50. Franco, C.; Antonow, M.B.; Beckenkamp, A.; Buffon, A.; Ceolin, T.; Tebaldi, M.L.; Silveira, G.P.; Stanisçuaski Guterres, S.; Raffin Pohlmann, A. PCL-b-P(MMA-co-DMAEMA)2 new triblock copolymer for novel pH-sensitive nanocapsules intended for drug delivery to tumors. React. Funct. Polym. 2017, 119, 116–124. [Google Scholar] [CrossRef]
  51. Shalumon, K.T.; Sheu, C.; Chen, C.-H.; Chen, S.-H.; Jose, G.; Kuo, C.-Y.; Chen, J.P. Multi-functional electrospun antibacterial core-shell nanofibrous membranes for prolonged prevention of post-surgical tendon adhesion and inflammation. Acta Biomater. 2018, 72, 121–136. [Google Scholar] [CrossRef]
  52. Kouhi, M.; Fathi, M.; Prabhakaran, M.P.; Shamanian, M.; Ramakrishna, S. Poly l lysine-modified phbv based nanofibrous scaffolds for bone cell mineralization and osteogenic differentiation. Appl. Surf. Sci. 2018, 457, 616–625. [Google Scholar] [CrossRef]
  53. Curry, E.J.; Le, T.T.; Das, R.; Ke, K.; Santorella, E.M.; Paul, D.; Chorsi, M.T.; Tran, K.T.M.; Baroody, J.; Borges, E.R.; et al. Biodegradable nanofiber-based piezoelectric transducer. Proc. Natl. Acad. Sci. USA 2020, 117, 214–220. [Google Scholar] [CrossRef] [PubMed]
  54. Goonoo, N.; Gimié, F.; Ait-Arsa, I.; Cordonin, C.; Andries, J.; Jhurrya, D.; Bhaw-Luximon, A. Piezoelectric core–shell PHBV/PDX blend scaffolds for reduced superficial wound contraction and scarless tissue regeneration. Biomater. Sci. 2021, 9, 5259–5274. [Google Scholar] [CrossRef] [PubMed]
  55. Keles, H.; Naylor, A.; Clegg, F.; Sammon, C. Investigation of factors influencing the hydrolytic degradation of single PLGA microparticles. Polym. Degrad. Stab. 2015, 119, 228–241. [Google Scholar] [CrossRef]
  56. Walker, J.; Albert, J.; Liang, D.; Sun, J.; Schutzman, R.; Kumar, R.; White, C.; Beck-Broichsitter, M.; Schwendeman, S. In Vitro Degradation and Erosion Behavior of Commercial PLGAs Used for Controlled Drug Delivery. Drug Deliv. Transl. Res. 2022, 13, 237–251. [Google Scholar] [CrossRef] [PubMed]
  57. Weng, Y.-X.; Wang, X.-L.; Wang, Y.-Z. Biodegradation Behavior of PHAs with Different Chemical Structures under Controlled Composting Conditions. Polym. Test. 2011, 30, 372–380. [Google Scholar] [CrossRef]
  58. Kovalcik, A.; Obruca, S.; Kalina, M.; Machovsky, M.; Enev, V.; Jakesova, M.; Sobkova, M.; Marova, I. Enzymatic Hydrolysis of Poly(3-Hydroxybutyrate-co-3-Hydroxyvalerate) Scaffolds. Materials 2020, 13, 2992. [Google Scholar] [CrossRef]
  59. Donate, R.; Monzón, M.; Alemán-Domínguez, M.E.; Ortega, Z. Enzymatic degradation study of PLA-based composite scaffolds. Rev. Adv. Mater. Sci. 2020, 59, 170–175. [Google Scholar] [CrossRef]
  60. Doyle, S.E.; Henry, L.; McGennisken, E.; Onofrillo, C.; Di Bella, C.; Duchi, S.; O’Connell, C.D.; Pirogova, E. Characterization of Polycaprolactone Nanohydroxyapatite Composites with Tunable Degradability Suitable for Indirect Printing. Polymers 2021, 13, 295. [Google Scholar] [CrossRef]
  61. Brdlík, P.; Borůvka, M.; Běhálek, L.; Lenfeld, P. The Influence of Additives and Environment on Biodegradation of PHBV Biocomposites. Polymers 2022, 14, 838. [Google Scholar] [CrossRef]
  62. Vaid, R.; Yildirim, E.; Pasquinelli, M.A.; King, M.W. Hydrolytic Degradation of Polylactic Acid Fibers as a Function of pH and Exposure Time. Molecules 2021, 26, 7554. [Google Scholar] [CrossRef]
  63. Leroux, A.; Nguyen, T.N.; Rangel, A.; Cacciapuoti, I.; Duprez, D.; Castner, D.G.; Migonney, V. Long-term hydrolytic degradation study of polycaprolactone films and fibers grafted with poly(sodium styrene sulfonate): Mechanism study and cell response. Biointerphases 2020, 15, 061006. [Google Scholar] [CrossRef] [PubMed]
  64. Polidar, M.; Metzsch-Zilligen, E.; Pfaendner, R. Controlled and Accelerated Hydrolysis of Polylactide (PLA) through Pentaerythritol Phosphites with Acid Scavengers. Polymers 2022, 14, 4237. [Google Scholar] [CrossRef] [PubMed]
  65. Chor, A.; Pires Gonçalves, R.; Machado Costa, A.; Farina, M.; Ponche, A.; Sirelli, L.; Schrodj, G.; Gree, S.; Rodrigues de Andrade, L.; Anselme, K.; et al. In Vitro Degradation of Electrospun Poly(Lactic-Co-Glycolic Acid) (PLGA) for Oral Mucosa Regeneration. Polymers 2020, 12, 1853. [Google Scholar] [CrossRef] [PubMed]
  66. Kobielarz, M.; Tomanik, M.; Mroczkowska, K.; Szustakiewicz, K.; Oryszczak, M.; Mazur, A. Laser-modified PLGA for implants: In vitro degradation and mechanical properties. Acta Bioeng. Biomech. 2020, 22, 179–192. [Google Scholar] [CrossRef]
  67. Rosato, A.; Romano, A.; Totaro, G.; Celli, A.; Fava, F.; Zanaroli, G.; Sisti, L. Enzymatic Degradation of the Most Common Aliphatic Bio-Polyesters and Evaluation of the Mechanisms Involved: An Extended Study. Polymers 2022, 14, 1850. [Google Scholar] [CrossRef]
  68. Dias, J.R.; Sousa, A.; Augusto, A.; Bártolo, P.J.; Granja, P.L. Electrospun Polycaprolactone (PCL) Degradation: An In Vitro and In Vivo Study. Polymers 2022, 14, 3397. [Google Scholar] [CrossRef]
  69. Shibata, A.; Yada, S.; Terakawa, M. Biodegradability of poly(lactic-co-glycolic acid) after femtosecond laser irradiation. Sci. Rep. 2016, 6, 27884. [Google Scholar] [CrossRef]
  70. Andhariy, J.A.; Jog, R.; Shen, J.; Choi, S.; Wang, Y.; Zou, Y.; Burgess, D.J. In vitro-in vivo correlation of parenteral PLGA microspheres: Effect of variable burst release. J. Control. Release 2019, 314, 25–37. [Google Scholar] [CrossRef]
  71. Abou Taleb, S.A.; Sobh, R.A.; Mourad, R.M. Investigating the Effect of Loading Curcuminoids Using PCL-PU-βCD Nano-Composites on Physico-Chemical Properties, In-Vitro Release, and Ex-Vivo Breast Cancer Cell-Line. Biointerface Res. Appl. Chem. 2022, 12, 4074–4102. [Google Scholar] [CrossRef]
  72. Fu, Z.; Li, L.; Wang, M.; Guo, X. Size control of drug nanoparticles stabilized by mPEG-b-PCL during flash nanoprecipitation. Colloid Polym. Sci. 2018, 296, 935–940. [Google Scholar] [CrossRef]
  73. Xu, J.; Chen, Y.; Jiang, X.; Gui, Z.; Zhang, L. Development of Hydrophilic Drug Encapsulation and Controlled Release Using a Modified Nanoprecipitation Method. Processes 2019, 7, 331. [Google Scholar] [CrossRef]
  74. Ge, M.; Sheng, Y.; Qi, S.; Cao, L.; Zhang, Y.; Yang, J. PLGA/chitosan–heparin composite microparticles prepared with microfluidics for the construction of hMSC aggregates. J. Mater. Chem. B 2020, 8, 9921–9932. [Google Scholar] [CrossRef]
  75. Walden, G.; Liao, X.; Riley, G.; Donell, S.; Raxworthy, M.R.; Saeed, A. Synthesis and Fabrication of Surface-Active Microparticles Using a Membrane Emulsion Technique and Conjugation of Model Protein via Strain-Promoted Azide–Alkyne Click Chemistry in Physiological Conditions. Bioconjug. Chem. 2019, 30, 531–535. [Google Scholar] [CrossRef] [PubMed]
  76. 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] [PubMed]
  77. Zhong, Y.; Godwin, P.; Jin, Y.; Xiao, H. Biodegradable polymers and green-based antimicrobial packaging materials: A mini-review. Adv. Ind. Eng. Polym. Res. 2020, 3, 27–35. [Google Scholar] [CrossRef]
  78. Jamaledin, R.; Sartorius, R.; Di Natale, C.; Vecchione, R.; De Berardinis, P.; Netti, P.A. Recombinant Filamentous Bacteriophages Encapsulated in Biodegradable Polymeric Microparticles for Stimulation of Innate and Adaptive Immune Responses. Microorganisms 2020, 8, 650. [Google Scholar] [CrossRef]
  79. Balakrishnan, K.; Casimeer, S.C.; Ghidan, A.Y.; Al Antary, T.M.; Singaravelu, A. Exploration of Antioxidant, Antibacterial Activities of Green Synthesized Hesperidin Loaded PLGA Nanoparticles. Biointerface Res. Appl. Chem. 2021, 11, 14520–14528. [Google Scholar] [CrossRef]
  80. Caciandone, M.; Niculescu, A.-G.; Grumezescu, V.; Bîrcă, A.C.; Ghica, I.C.; Vasile, B.Ș.; Oprea, O.; Nica, I.C.; Stan, M.S.; Holban, A.M.; et al. Magnetite Nanoparticles Functionalized with Therapeutic Agents for Enhanced ENT Antimicrobial Properties. Antibiotics 2022, 11, 623. [Google Scholar] [CrossRef]
  81. Hasan, N.; Cao, J.; Lee, J.; Hlaing, S.P.; Oshi, M.A.; Naeem, M.; Ki, M.-H.; Lee, B.L.; Jung, Y.; Yoo, J.-W. Bacteria-Targeted Clindamycin Loaded Polymeric Nanoparticles: Effect of Surface Charge on Nanoparticle Adhesion to MRSA, Antibacterial Activity, and Wound Healing. Pharmaceutics 2019, 11, 236. [Google Scholar] [CrossRef]
  82. Da Costa, D.; Exbrayat-Héritier, C.; Rambaud, B.; Megy, S.; Terreux, R.; Verrier, B.; Primard, C. Surface charge modulation of rifampicin-loaded PLA nanoparticles to improve antibiotic delivery in Staphylococcus aureus biofilms. J. Nanobiotechnol. 2021, 19, 12. [Google Scholar] [CrossRef]
  83. Ranjanamala, T.; Vanmathiselvi, K.; Casimeer, S.G.; Ghidan, A.Y. Synthesis and Characterization of Dose-Dependent Antioxidants and Antimicrobial Activit.y of Phloretin Loaded PLGA Nanoparticles. Biointerface Res. Appl. Chem. 2022, 12, 3076–3089. [Google Scholar] [CrossRef]
  84. Liu, Y.; Busscher, H.J.; Zhao, B.; Li, Y.; Zhang, Z.; van der Mei, H.C.; Ren, Y.; Shi, L. Surface-Adaptive, Antimicrobially Loaded, Micellar Nanocarriers with Enhanced Penetration and Killing Efficiency in Staphylococcal Biofilms. ACS Nano 2016, 10, 4779–4789. [Google Scholar] [CrossRef] [PubMed]
  85. Wan, F.; Bohr, S.S.-R.; Kłodzińska, S.N.; Jumaa, H.; Huang, Z.; Nylander, T.; Thygesen, M.B.; Sørensen, K.K.; Jensen, K.J.; Sternberg, C.; et al. Ultrasmall TPGS–PLGA Hybrid Nanoparticles for Site-Specific Delivery of Antibiotics into Pseudomonas aeruginosa Biofilms in Lungs. ACS Appl. Mater. Interfaces 2020, 12, 380–389. [Google Scholar] [CrossRef]
  86. Brauner, B.; Semmler, J.; Rauch, D.; Nokaj, M.; Haiss, P.; Schwarz, P.; Wirth, M.; Gabor, F. Trimethoprim-Loaded PLGA Nanoparticles Grafted with WGA as Potential Intravesical Therapy of Urinary Tract Infections—Studies on Adhesion to SV-HUCs Under Varying Time, pH, and Drug-Loading Conditions. ACS Omega 2020, 5, 17377–17384. [Google Scholar] [CrossRef]
  87. Torge, A.; Wagner, S.; Chaves, P.S.; Oliveira, E.G.; Guterres, S.S.; Pohlmann, A.R.; Titz, A.; Schneider, M.; Beck, R.C.R. Ciprofloxacin-loaded lipid-core nanocapsules as mucus penetrating drug delivery system intended for the treatment of bacterial infections in cystic fibrosis. Int. J. Pharm. 2017, 527, 92–102. [Google Scholar] [CrossRef]
  88. Mayank, H.; Sharma, A.; Verma, R.K.; Shukla, R. Polycaprolactone based nano-carrier for co-administration of moxifloxacin and rutin and its In-vitro evaluation for sepsis. J. Drug Deliv. Sci. Technol. 2019, 54, 101286. [Google Scholar] [CrossRef]
  89. Casciaro, B.; d’Angelo, I.; Zhang, X.; Loffredo, M.R.; Conte, G.; Cappiello, F.; Quaglia, F.; Di, Y.P.; Ungaro, F.; Mangoni, M.L. Poly(lactide-co-glycolide) Nanoparticles for Prolonged Therapeutic Efficacy of Esculentin-1a-Derived Antimicrobial Peptides against Pseudomonas aeruginosa Lung Infection: In Vitro and in Vivo Studies. Biomacromolecules 2019, 20, 1876–1888. [Google Scholar] [CrossRef] [PubMed]
  90. Englert, C.; Brendel, J.C.; Majdanski, T.C.; Yildirim, T.; Schubert, S.; Gottschaldt, M.; Windhab, N.; Schubert, U.S. Pharmapolymers in the 21st century: Synthetic polymers in drug delivery applications. Prog. Polym. Sci. 2018, 87, 107–164. [Google Scholar] [CrossRef]
  91. Nofar, M.; Sacligil, D.; Carreau, P.J.; Kamal, M.R.; Heuzey, M.C. Poly (lactic acid) blends: Processing, properties and applications. Int. J. Biol. Macromol. 2019, 125, 307–360. [Google Scholar] [CrossRef] [PubMed]
  92. Lee, B.K.; Yun, Y.; Park, K. PLA micro- and nano-particles. Adv. Drug Deliv. Rev. 2016, 107, 176–191. [Google Scholar] [CrossRef] [Green Version]
  93. Jain, A.; Kunduru, K.R.; Basu, A.; Mizrahi, B.; Domb, A.J.; Khan, W. Injectable formulations of poly(lactic acid) and its copolymers in clinical use. Adv. Drug Deliv. Rev. 2016, 107, 213–227. [Google Scholar] [CrossRef] [PubMed]
  94. Silva, C.; Yudice, E.; Campini, P.; Rosa, D. The performance evaluation of Eugenol and Linalool microencapsulated by PLA on their activities against pathogenic bacteria. Mater. Today Chem. 2021, 21, 100493. [Google Scholar] [CrossRef]
  95. Niza, E.; Božik, M.; Bravo, I.; Clemente-Casares, P.; Lara-Sanchez, A.; Juan, A.; Klouček, P.; Alonso-Moreno, C. PEI-coated PLA nanoparticles to enhance the antimicrobial activity of carvacrol. Food Chem. 2020, 328, 127131. [Google Scholar] [CrossRef] [PubMed]
  96. Sabee, M.M.; Awang, M.; Bustami, Y.; Hamid, Z.A.A. Antimicrobial activity evaluation for gentamicin loaded PLA microspheres. Materials Today. Proceedings 2019, 16, 2060–2066. [Google Scholar] [CrossRef]
  97. Chinavinijkul, P.; Riansuwan, K.; Kiratisin, P.; Srisang, S.; Nasongkla, N. Dip-and Spray-coating of Schanz pin with PLA and PLA nanosphere for prolonged antibacterial activity. J. Drug Deliv. Sci. Technol. 2021, 65, 102667. [Google Scholar] [CrossRef]
  98. Ding, D.; Zhu, Q. Recent advances of PLGA micro/nanoparticles for the delivery of biomacromolecular therapeutics. Mater. Sci. Eng. C Mater. Biol. Appl. 2018, 92, 1041–1060. [Google Scholar] [CrossRef] [PubMed]
  99. Swider, E.; Koshkina, O.; Tel, J.; Cruz, L.J.; de Vries, I.J.M.; Srinivas, M. Customizing poly(lactic-co-glycolic acid) particles for biomedical applications. Acta Biomater. 2018, 73, 38–51. [Google Scholar] [CrossRef]
  100. Vilos, C.; Velasquez, L.A.; Rodas, P.I.; Zepeda, K.; Bong, S.J.; Herrera, N.; Cantin, M.; Simon, F.; Constandil, L. Preclinical development and in vivo efficacy of ceftiofur-plga microparticles. PLoS ONE 2015, 10, e0123335. [Google Scholar] [CrossRef]
  101. Singh, Y.; Srinivas, A.; Gangwar, M.; Meher, J.G.; Misra-Bhattacharya, S.; Chourasia, M.K. Subcutaneously administered ultrafine plga nanoparticles containing doxycycline hydrochloride target lymphatic filarial parasites. Mol. Pharm. 2016, 13, 2084–2094. [Google Scholar] [CrossRef]
  102. Ali, M.; Walboomers, X.F.; Jansen, J.A.; Yang, F. Influence of formulation parameters on encapsulation of doxycycline in plga microspheres prepared by double emulsion technique for the treatment of periodontitis. J. Drug Deliv. Sci. Technol. 2019, 52, 263–271. [Google Scholar] [CrossRef]
  103. Wang, F.; Wang, M.; She, Z.; Fan, K.; Xu, C.; Chu, B.; Chen, C.; Shi, S.; Tan, R. Collagen/chitosan based two-compartment and bi-functional dermal scaffolds for skin regeneration. Mater. Sci. Eng. C Mater. Biol. Appl. 2015, 52, 155–162. [Google Scholar] [CrossRef] [PubMed]
  104. Fan, Y.; Zheng, X.; Ali, Y.; Berggren, P.O.; Loo, S.C.J. Local release of rapamycin by microparticles delays islet rejection within the anterior chamber of the eye. Sci. Rep. 2019, 9, 3918. [Google Scholar] [CrossRef] [PubMed]
  105. Wu, J.; Zuo, Y.; Hu, Y.; Wang, J.; Li, J.; Qiao, B.; Jiang, D. Development and in vitro characterization of drug delivery system of rifapentine for osteoarticular tuberculosis. Drug Des. Dev. Ther. 2015, 9, 1359–1366. [Google Scholar] [CrossRef] [PubMed]
  106. Huang, J.; Chen, Z.; Li, Y.; Li, L.; Zhang, G. Rifapentine-linezolid-loaded plga microspheres for interventional therapy of cavitary pulmonary tuberculosis: Preparation and in vitro characterization. Drug Des. Dev. Ther. 2017, 11, 585–592. [Google Scholar] [CrossRef]
  107. Cong, Y.; Quan, C.; Liu, M.; Liu, J.; Huang, G.; Tong, G.; Yin, Y.; Zhang, C.; Jiang, Q. Alendronate-decorated biodegradable polymeric micelles for potential bone-targeted delivery of vancomycin. J. Biomater. Sci. Polym. Ed. 2015, 26, 629–643. [Google Scholar] [CrossRef]
  108. Hill, M.; Cunningham, R.N.; Hathout, R.M.; Johnston, C.; Hardy, J.G.; Migaud, M.E. Formulation of antimicrobial tobramycin loaded PLGA nanoparticles via complexation with AOT. J. Funct. Biomater. 2019, 10, 26. [Google Scholar] [CrossRef]
  109. Rancan, F.; Jurisch, J.; Günday, C.; Türeli, E.; Blume-Peytavi, U.; Vogt, A.; Schaudinn, C.; Günday-Türeli, N. Screening of surfactants for improved delivery of antimicrobials and poly-lactic-co-glycolic acid particles in wound tissue. Pharmaceutics 2021, 13, 1093. [Google Scholar] [CrossRef]
  110. Karp, F.; Turino, L.N.; Helbling, I.M.; Islan, G.A.; Luna, J.A.; Estenoz, D.A. In situ Formed Implants, Based on PLGA and Eudragit Blends, for Novel Florfenicol Controlled Release Formulations. J. Pharm. Sci. 2021, 110, 1270–1278. [Google Scholar] [CrossRef]
  111. Yurtdaş-Kırımlıoğlu, G.; Görgülü, Ş. Surface modification of PLGA nanoparticles with chitosan or Eudragit® RS 100: Characterization, prolonged release, cytotoxicity, and enhanced antimicrobial activity. J. Drug Deliv. Sci. Technol. 2021, 61, 102145. [Google Scholar] [CrossRef]
  112. Wang, Y.; Guo, L.; Liu, J.; Huang, X.; Wang, X.; Guo, X.; You, X.; Li, W.; Li, L.; Sun, T.; et al. Chitosan/PLGA shell nanoparticles as Tylotoin delivery platform for advanced wound healing. Int. J. Biol. Macromol. 2022, 220, 395–405. [Google Scholar] [CrossRef]
  113. Almukainzi, M.; El-Masry, T.A.; Negm, W.A.; Elekhnawy, E.; Saleh, A.; Sayed, E.A.; Khattab, M.A.; Abdelkader, D.H. Gentiopicroside PLGA Nanospheres: Fabrication, in vitro Characterization, Antimicrobial Action, and in vivo Effect for Enhancing Wound Healing in Diabetic Rats. Int. J. Nanomed. 2022, 17, 1203–1225. [Google Scholar] [CrossRef] [PubMed]
  114. Pola, C.C.; Moraes, A.R.F.; Medeiros, E.A.A.; Teófilo, R.F.; Soares, N.F.F.; Gomes, C.L. Development and optimization of pH-responsive PLGA-chitosan nanoparticles for triggered release of antimicrobials. Food Chem. 2019, 295, 671–679. [Google Scholar] [CrossRef] [PubMed]
  115. Cheng, Y.; Qin, J.; Huang, Y.; Wang, T. The antimicrobial effects of PLGA microspheres containing the antimicrobial peptide OP-145 on clinically isolated pathogens in bone infections. Sci. Rep. 2022, 12, 14541. [Google Scholar] [CrossRef] [PubMed]
  116. Li, Y.; Na, R.; Wang, X.; Liu, H.; Zhao, L.; Sun, X.; Ma, G.; Cui, F. Fabrication of antimicrobial peptide-loaded PLGA/Chitosan composite microspheres for long-Acting bacterial resistance. Molecules 2017, 22, 1637. [Google Scholar] [CrossRef]
  117. Sousa, F.F.O.; Nojosa, J.S.; Alencar, C.A.A.; Alcantara, A.P.M.P.; Araújo, R.S.; Yamauti, M.; Rodrigues, L.K.A. Design and characterization of digluconate and diacetate chlorhexidine loaded-PLGA microparticles for dental applications. J. Drug Deliv. Sci. Technol. 2021, 62, 102361. [Google Scholar] [CrossRef]
  118. Akram, Z.; Daood, U.; Aati, S.; Ngo, H.; Fawzy, A.S. Formulation of pH-sensitive chlorhexidine-loaded/mesoporous silica nanoparticles modified experimental dentin adhesive. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 122, 111894. [Google Scholar] [CrossRef]
  119. Mahmoud, M.Y.; Steinbach-Rankins, J.M.; Demuth, D.R. Functional assessment of peptide-modified PLGA nanoparticles against oral biofilms in a murine model of periodontitis. J. Control. Release 2019, 297, 3–13. [Google Scholar] [CrossRef]
  120. Agarwal, R.; Johnson, C.T.; Imhoff, B.R.; Donlan, R.M.; McCarty, N.A.; García, A.J. Inhaled bacteriophage-loaded polymeric microparticles ameliorate acute lung infections. Nat. Biomed. Eng. 2018, 2, 841–849. [Google Scholar] [CrossRef]
  121. Anversa Dimer, F.; de Souza Carvalho-Wodarz, C.; Goes, A.; Cirnski, K.; Herrmann, J.; Schmitt, V.; Pätzold, L.; Abed, N.; De Rossi, C.; Bischoff, M.; et al. PLGA nanocapsules improve the delivery of clarithromycin to kill intracellular Staphylococcus aureus and Mycobacterium abscessus. Nanomed. Nanotechnol. Biol. Med. 2020, 24, 102125. [Google Scholar] [CrossRef]
  122. Abdelghany, S.; Parumasivam, T.; Pang, A.; Roediger, B.; Tang, P.; Jahn, K.; Britton, W.J.; Chan, H.K. Alginate modified-PLGA nanoparticles entrapping amikacin and moxifloxacin as a novel host-directed therapy for multidrug-resistant tuberculosis. J. Drug Deliv. Sci. Technol. 2019, 52, 642–651. [Google Scholar] [CrossRef]
  123. Lababidi, N.; Montefusco-Pereira, C.V.; de Souza Carvalho-Wodarz, C.; Lehr, C.M.; Schneider, M. Spray-dried multidrug particles for pulmonary co-delivery of antibiotics with N-acetylcysteine and curcumin-loaded PLGA-nanoparticles. Eur. J. Pharm. Biopharm. 2020, 157, 200–210. [Google Scholar] [CrossRef] [PubMed]
  124. De Ávila, B.E.F.; Angsantikul, P.; Li, J.; Angel Lopez-Ramirez, M.; Ramírez-Herrera, D.E.; Thamphiwatana, S.; Chen, C.; Delezuk, J.; Samakapiruk, R.; Ramez, V.; et al. Micromotor-enabled active drug delivery for in vivo treatment of stomach infection. Nat. Commun. 2017, 8, 272. [Google Scholar] [CrossRef] [PubMed]
  125. Luo, M.; Jia, Y.Y.; Jing, Z.W.; Li, C.; Zhou, S.Y.; Mei, Q.B.; Zhang, B.L. Construction and optimization of pH-sensitive nanoparticle delivery system containing PLGA and UCCs-2 for targeted treatment of Helicobacter pylori. Colloids Surf. B Biointerfaces 2018, 164, 11–19. [Google Scholar] [CrossRef] [PubMed]
  126. Raza, A.; Miles, J.A.; Sime, F.B.; Ross, B.P.; Roberts, J.A.; Popat, A.; Kumeria, T.; Falconer, J.R. PLGA encapsulated γ-cyclodextrin-meropenem inclusion complex formulation for oral delivery. Int. J. Pharm. 2021, 597, 120280. [Google Scholar] [CrossRef] [PubMed]
  127. Mahboob, T.; Nawaz, M.; de Lourdes Pereira, M.; Tian-Chye, T.; Samudi, C.; Sekaran, S.D.; Wiart, C.; Nissapatorn, V. PLGA nanoparticles loaded with Gallic acid- a constituent of Leea indica against Acanthamoeba triangularis. Sci. Rep. 2020, 10, 8954. [Google Scholar] [CrossRef]
  128. Bao, Q.; Zhang, Z.; Yu, B.; Sun, H.; Leung, P.H.; Tao, X. Synthesis of Polylactic Acid Oligomers for Broad-Spectrum Antimicrobials. Polymers 2022, 14, 4399. [Google Scholar] [CrossRef] [PubMed]
  129. Demchenko, V.; Mamunya, Y.; Kobylinskyi, S.; Riabov, S.; Naumenko, K.; Zahorodnia, S.; Povnitsa, O.; Rybalchenko, N.; Iurzhenko, M.; Adamus, G.; et al. Structure-Morphology-Antimicrobial and Antiviral Activity Relationship in Silver-Containing Nanocomposites Based on Polylactide. Molecules 2022, 27, 3769. [Google Scholar] [CrossRef] [PubMed]
  130. Meena, J.; Singhvi, P.; Srichandan, S.; Dandotiya, J.; Verma, J.; Singh, M.; Ahuja, R.; Panwar, N.; Wani, T.Q.; Khatri, R.; et al. RBD decorated PLA nanoparticle admixture with aluminum hydroxide elicit robust and long lasting immune response against SARS-CoV-2. Eur. J. Pharm. Biopharm. 2022, 176, 43–53. [Google Scholar] [CrossRef] [PubMed]
  131. Pourhajibagher, M.; Azimi, M.; Haddadi-Asl, V.; Ahmadi, H.; Gholamzad, M.; Ghorbanpour, S.; Bahador, A. Robust antimicrobial photodynamic therapy with curcumin-poly (lactic-co-glycolic acid) nanoparticles against COVID-19: A preliminary in vitro study in Vero cell line as a model. Photodiagn. Photodyn. Ther. 2021, 34, 102286. [Google Scholar] [CrossRef]
  132. Yurtdaş-Kırımlıoğlu, G.; Görgülü, Ş.; Güleç, K.; Kıyan, H.T. Nanoarchitectonics of PLGA based polymeric nanoparticles with oseltamivir phosphate for lung cancer therapy: In vitro-in vivo evaluation. J. Drug Deliv. Sci. Technol. 2022, 67, 102996. [Google Scholar] [CrossRef]
  133. Dhoke, D.M.; Basaiyye, S.S.; Khedekar, P.B. Development and characterization of L-HSA conjugated PLGA nanoparticle for hepatocyte targeted delivery of antiviral drug. J. Drug Deliv. Sci. Technol. 2018, 47, 77–94. [Google Scholar] [CrossRef]
  134. Ayoub, M.M.; Elantouny, N.G.; El-Nahas, H.M.; Ghazy, F.E.D.S. Injectable PLGA Adefovir microspheres; the way for long term therapy of chronic hepatitis-B. Eur. J. Pharm. Sci. 2018, 118, 24–31. [Google Scholar] [CrossRef]
  135. Fleitas-Salazar, N.; Lamazares, E.; Pedroso-Santana, S.; Kappes, T.; Pérez-Alonso, T.; Hidalgo, A.; Altamirano, C.; Sánchez, O.; Fernández, K.; Toledo, J. Long-term release of bioactive interferon-alpha from PLGA-chitosan microparticles: In vitro and in vivo studies. Biomater. Adv. 2022, 143, 213167. [Google Scholar] [CrossRef] [PubMed]
  136. MacKerracher, A.; Sommershof, A.; Groettrup, M. PLGA particle vaccination elicits resident memory CD8 T cells protecting from tumors and infection. Eur. J. Pharm. Sci. 2022, 175, 106209. [Google Scholar] [CrossRef] [PubMed]
  137. Tukhvatulin, A.; Dzharullaeva, A.; Erokhova, A.; Zemskaya, A.; Balyasin, M.; Ozharovskaia, T.; Zubkova, O.; Shevlyagina, N.; Zhukhovitsky, V.; Fedyakina, I.; et al. Adjuvantation of an Influenza Hemagglutinin Antigen with TLR4 and NOD2 Agonists Encapsulated in Poly(D,L-Lactide-Co-Glycolide) Nanoparticles Enhances Immunogenicity and Protection against Lethal Influenza Virus Infection in Mice. Vaccines 2020, 8, 519. [Google Scholar] [CrossRef] [PubMed]
  138. Kingstad-Bakke, B.; Toy, R.; Lee, W.; Pradhan, P.; Vogel, G.; Marinaik, C.B.; Larsen, A.; Gates, D.; Luu, T.; Pandey, B.; et al. Polymeric Pathogen-Like Particles-Based Combination Adjuvants Elicit Potent Mucosal T Cell Immunity to Influenza A Virus. Front. Immunol. 2021, 11, 559382. [Google Scholar] [CrossRef] [PubMed]
  139. Sanna, V.; Satta, S.; Hsiai, T.; Sechi, M. Development of targeted nanoparticles loaded with antiviral drugs for SARS-CoV-2 inhibition. Eur. J. Med. Chem. 2022, 231, 114121. [Google Scholar] [CrossRef]
  140. Bemdos Santos, R.; Funguetto-Ribeiro, A.C.; Ramos Maciel, T.; Pereira Fonseca, D.; Reis Favarin, F.; Rubert Nogueira-Librelotto, D.; Gomes de Gomes, M.; Ueda Nakamura, T.; Bueno Rolim, C.M.; Haas, S.E. In vivo and in vitro per se effect evaluation of Polycaprolactone and Eudragit® RS100-based nanoparticles. Biomed. Pharmacother. 2022, 153, 113410. [Google Scholar] [CrossRef]
  141. Varan, C.; Wickström, H. l Sandler, N.; Aktaş, Y.; Bilensoy, E. Inkjet printing of antiviral PCL nanoparticles and anticancer cyclodextrin inclusion complexes on bioadhesive film for cervical administration. Int. J. Pharm. 2017, 531, 701–713. [Google Scholar] [CrossRef]
  142. Varan, C.; Şen, M.; Sandler, N.; Aktaş, Y.; Bilensoy, E. Mechanical characterization and ex vivo evaluation of anticancer and antiviral drug printed bioadhesive film for the treatment of cervical cancer. Eur. J. Pharm. Sci. 2019, 130, 114–123. [Google Scholar] [CrossRef]
  143. Zhou, L.; Li, A.; Wang, H.; Sun, W.; Zuo, S.; Li, C. Preparation and characterization of luteolin-loaded MPEG-PCL-g-PEI micelles for oral Candida albicans infection. J. Drug Deliv. Sci. Technol. 2021, 63, 102454. [Google Scholar] [CrossRef]
  144. Shi, Q.; Daisy, E.R.A.C.; Yang, G.; Zhang, J.; Mickymaray, S.; Alfaiz, F.A.; Paramasivam, A.; Rajan, M. Multifeatured guar gum armed drug delivery system for the delivery of ofloxacin drug to treat ophthalmic dieases. Arab. J. Chem. 2021, 14, 103118. [Google Scholar] [CrossRef]
  145. Javaid, S.; Ahmad, N.M.; Mahmood, A.; Nasir, H.; Iqbal, M.; Ahmad, N.; Irshad, S. Cefotaxime Loaded Polycaprolactone Based Polymeric Nanoparticles with Antifouling Properties for In-Vitro Drug Release Applications. Polymers 2021, 13, 2180. [Google Scholar] [CrossRef]
  146. Srisang, S.; Nasongkla, N. Spray coating of foley urinary catheter by chlorhexidine-loadedpoly(ε-caprolactone) nanospheres: Effect of lyoprotectants, characteristics, and antibacterial activity evaluation. Pharm. Dev. Technol. 2019, 24, 402–409. [Google Scholar] [CrossRef] [PubMed]
  147. Srisang, S.; Nasongkla, N. Layer-by-layer dip coating of Foley urinary catheters by chlorhexidine-loaded micelles. J. Drug Deliv. Sci. Technol. 2019, 49, 235–242. [Google Scholar] [CrossRef]
  148. Srisang, S.; Boongird, A.; Ungsurungsie, M.; Wanasawas, P.; Nasongkla, N. In vivo catheterization study of chlorhexidine-loaded nanoparticle coated Foley urinary catheters in male New Zealand white rabbits. J. Biomed. Mater. Res.—Part B Appl. Biomater. 2021, 109, 1836–1843. [Google Scholar] [CrossRef]
  149. Amaro, L.; Correia, D.M.; Martins, P.M.; Botelho, G.; Carabineiro, S.A.C.; Ribeiro, C.; Lanceros-Mendez, S. Morphology dependence degradation of electro-and magnetoactive poly(3-hydroxybutyrateco-hydroxyvalerate) for tissue engineering applications. Polymers 2020, 12, 953. [Google Scholar] [CrossRef]
  150. Álvarez-Álvarez, L.; Barral, L.; Bouza, R.; Farrag, Y.; Otero-Espinar, F.; Feijóo-Bandín, S.; Lago, F. Hydrocortisone loaded poly-(3-hydroxybutyrate-co-3-hydroxyvalerate) nanoparticles for topical ophthalmic administration: Preparation, characterization and evaluation of ophthalmic toxicity. Int. J. Pharm. 2019, 568, 118519. [Google Scholar] [CrossRef]
  151. Wu, M.; Zhong, C.; Zhang, Q.; Wang, L.; Wang, L.; Liu, Y.; Zhang, X.; Zhao, X. pH-responsive delivery vehicle based on RGD-modified polydopamine-paclitaxel-loaded poly (3-hydroxybutyrate-co-3-hydroxyvalerate) nanoparticles for targeted therapy in hepatocellular carcinoma. J. Nanobiotechnol. 2021, 19, 39. [Google Scholar] [CrossRef]
  152. Vilos, C.; Constandil, L.; Rodas, P.I.; Cantin, M.; Zepeda, K.; Herrera, N.; Velasquez, L.A. Evaluation of ceftiofur-PHBV microparticles in rats. Drug Des. Dev. Ther. 2014, 8, 651–666. [Google Scholar] [CrossRef] [Green Version]
  153. Solar, P.; González, G.; Vilos, C.; Herrera, N.; Juica, N.; Moreno, M.; Simon, F.; Velásquez, L. Multifunctional polymeric nanoparticles doubly loaded with SPION and ceftiofur retain their physical and biological properties. J. Nanobiotechnol. 2015, 13, 14. [Google Scholar] [CrossRef] [PubMed]
  154. Li, W.; Jan, Z.; Ding, Y.; Liu, Y.; Janko, C.; Pischetsrieder, M.; Alexiou, C.; Boccaccini, A.R. Facile preparation of multifunctional superparamagnetic PHBV microspheres containing SPIONs for biomedical applications. Sci. Rep. 2016, 6, 23140. [Google Scholar] [CrossRef] [PubMed]
  155. Idris, M.I.; Zaloga, J.; Detsch, R.; Roether, J.A.; Unterweger, H.; Alexiou, C.; Boccaccini, A.R. Surface Modification of SPIONs in PHBV Microspheres for Biomedical Applications. Sci. Rep. 2018, 8, 7286. [Google Scholar] [CrossRef] [PubMed]
  156. Perveen, K.; Masood, F.; Hameed, A. Preparation, characterization and evaluation of antibacterial properties of epirubicin loaded PHB and PHBV nanoparticles. Int. J. Biol. Macromol. 2020, 144, 259–266. [Google Scholar] [CrossRef]
  157. Chen, Q.; Li, W.; Goudouri, O.M.; Ding, Y.; Cabanas-Polo, S.; Boccaccini, A.R. Electrophoretic deposition of antibiotic loaded PHBV microsphere-alginate composite coating with controlled delivery potential. Colloids Surf. B Biointerfaces 2015, 130, 199–206. [Google Scholar] [CrossRef] [PubMed]
  158. Chotchindakun, K.; Pekkoh, J.; Ruangsuriya, J.; Zheng, K.; Unalan, I.; Boccaccini, A.R. Fabrication and characterization of cinnamaldehyde-loaded mesoporous bioactive glass nanoparticles/PHBV-based microspheres for preventing bacterial infection and promoting bone tissue regeneration. Polymers 2021, 13, 1794. [Google Scholar] [CrossRef] [PubMed]
  159. Li, W.; Ding, Y.; Rai, R.; Roether, J.A.; Schubert, D.W.; Boccaccini, A.R. Preparation and characterization of PHBV microsphere/45S5 bioactive glass composite scaffolds with vancomycin releasing function. Mater. Sci. Eng. C Mater. Biol. Appl. 2014, 41, 320–328. [Google Scholar] [CrossRef] [PubMed]
  160. Shalom, H.; Sui, X.; Elianov, O.; Brumfeld, V.; Rosentsveig, R.; Pinkas, I.; Feldman, Y.; Kampf, N.; Wagner, H.D.; Lachman, N.; et al. Nanocomposite of Poly(l-Lactic Acid) with Inorganic Nanotubes of WS2. Lubricants 2019, 7, 28. [Google Scholar] [CrossRef]
  161. Escobar, A.; Muzzio, N.; Moya, S.E. Antibacterial Layer-by-Layer Coatings for Medical Implants. Pharmaceutics 2021, 13, 16. [Google Scholar] [CrossRef]
  162. Liao, C.; Li, Y.; Tjong, S.C. Antibacterial Activities of Aliphatic Polyester Nanocomposites with Silver Nanoparticles and/or Graphene Oxide Sheets. Nanomaterials 2019, 9, 1102. [Google Scholar] [CrossRef] [Green Version]
  163. Mir, M.; Ali, M.N.; Barakullah, A.; Gulzar, A.; Arshad, M.; Fatima, S.; Asad, M. Synthetic polymeric biomaterials for wound healing: A review. Prog. Biomater. 2018, 7, 1–21. [Google Scholar] [CrossRef] [PubMed]
  164. Akshaya, S.; Rowlo, P.K.; Dukle, A.; Nathanael, A.J. Antibacterial Coatings for Titanium Implants: Recent Trends and Future Perspectives. Antibiotics 2022, 11, 1719. [Google Scholar] [CrossRef] [PubMed]
  165. Kravanja, K.A.; Finšgar, M. A review of techniques for the application of bioactive coatings on metal-based implants to achieve controlled release of active ingredients. Mater. Des. 2022, 217, 110653. [Google Scholar] [CrossRef]
  166. Pušnik Črešnar, K.; Aulova, A.; Bikiaris, D.N.; Lambropoulou, D.; Kuzmič, K.; Fras Zemljič, L. Incorporation of Metal-Based Nanoadditives into the PLA Matrix: Effect of Surface Properties on Antibacterial Activity and Mechanical Performance of PLA Nanoadditive Films. Molecules 2021, 26, 4161. [Google Scholar] [CrossRef] [PubMed]
  167. Zhu, J.-Y.; Tang, C.-H.; Yin, S.-W.; Yang, X.-Q. Development and characterization of novel antimicrobial bilayer films based on Polylactic acid (PLA)/Pickering emulsions. Carbohydr. Polym. 2018, 181, 727–735. [Google Scholar] [CrossRef]
  168. Gherasim, O.; Grumezescu, A.M.; Grumezescu, V.; Iordache, F.; Vasile, B.S.; Holban, A.M. Bioactive Surfaces of Polylactide and Silver Nanoparticles for the Prevention of Microbial Contamination. Materials 2020, 13, 768. [Google Scholar] [CrossRef]
  169. Gherasim, O.; Popescu, R.C.; Grumezescu, V.; Mogoșanu, G.D.; Mogoantă, L.; Iordache, F.; Holban, A.M.; Vasile, B.S.; Bîrcă, A.C.; Oprea, O.-C.; et al. MAPLE Coatings Embedded with Essential Oil-Conjugated Magnetite for Anti-Biofilm Applications. Materials 2021, 14, 1612. [Google Scholar] [CrossRef]
  170. Macha, I.J.; Cazalbou, S.; Ben-Nissan, B.; Harvey, K.L.; Milthorpe, B. Marine structure derived calcium phosphate–polymer biocomposites for local antibiotic delivery. Mar. Drugs 2015, 13, 666–680. [Google Scholar] [CrossRef]
  171. Karacan, I.; Ben-Nissan, B.; Wang, H.A.; Juritza, A.; Swain, M.V.; Müller, W.H.; Chou, J.; Stamboulis, A.; Macha, I.J.; Taraschi, V. Mechanical testing of antimicrobial biocomposite coating on metallic medical implants as drug delivery system. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 104, 109757. [Google Scholar] [CrossRef]
  172. Grumezescu, V.; Socol, G.; Grumezescu, A.M.; Holban, A.M.; Ficai, A.; Truşcǎ, R.; Bleotu, C.; Balaure, P.C.; Cristescu, R.; Chifiriuc, M.C. Functionalized antibiofilm thin coatings based on PLA-PVA microspheres loaded with usnic acid natural compounds fabricated by MAPLE. Appl. Surf. Sci. 2014, 302, 262–267. [Google Scholar] [CrossRef]
  173. Li, Z.; Liu, X.; Li, Y.; Lan, X.; Leung, P.H.; Li, J.; Li, G.; Xie, M.; Han, Y.; Lin, X. Composite membranes of recombinant silkworm antimicrobial peptide and poly (L-lactic acid)(PLLA) for biomedical application. Sci. Rep. 2016, 6, 1–12. [Google Scholar] [CrossRef] [PubMed]
  174. Sardareh, E.A.; Shahzeidi, M.; Ardestani, M.T.S.; Mousavi-Khattat, M.; Zarepour, A.; Zarrabi, A. Antimicrobial Activity of Blow Spun PLA/Gelatin Nanofibers Containing Green Synthesized Silver Nanoparticles against Wound Infection-Causing Bacteria. Bioengineering 2022, 9, 518. [Google Scholar] [CrossRef] [PubMed]
  175. Bardania, H.; Mahmoudi, R.; Bagheri, H.; Salehpour, Z.; Fouani, M.H.; Darabian, B.; Khoramrooz, S.S.; Mousavizadeh, A.; Kowsari, M.; Moosavifard, S.E. Facile preparation of a novel biogenic silver-loaded Nanofilm with intrinsic anti-bacterial and oxidant scavenging activities for wound healing. Sci. Rep. 2020, 10, 1–14. [Google Scholar] [CrossRef] [PubMed]
  176. Ferreira, K.N.; Oliveira, R.R.; Castellano, L.R.C.; Bonan, P.R.F.; Carvalho, O.V.; Pena, L.; Souza, J.R.; Oliveira, J.E.; Medeiros, E.S. Controlled release and antiviral activity of acyclovir-loaded PLA/PEG nanofibers produced by solution blow spinning. Biomater. Adv. 2022, 136, 212785. [Google Scholar] [CrossRef]
  177. Cerro, D.; Bustos, G.; Villegas, C.; Buendia, N.; Truffa, G.; Godoy, M.P.; Rodriguez, F.; Rojas, A.; Galotto, M.J.; Constandil, L. Effect of supercritical incorporation of cinnamaldehyde on physical-chemical properties, disintegration and toxicity studies of PLA/lignin nanocomposites. Int. J. Biol. Macromol. 2021, 167, 255–266. [Google Scholar] [CrossRef]
  178. Zodrow, K.R.; Schiffman, J.D.; Elimelech, M. Biodegradable polymer (PLGA) coatings featuring cinnamaldehyde and carvacrol mitigate biofilm formation. Langmuir 2012, 28, 13993–13999. [Google Scholar] [CrossRef]
  179. Garcia-Orue, I.; Gainza, G.; Garcia-Garcia, P.; Gutierrez, F.B.; Aguirre, J.J.; Hernandez, R.M.; Delgado, A.; Igartua, M. Composite nanofibrous membranes of PLGA/Aloe vera containing lipid nanoparticles for wound dressing applications. Int. J. Pharm. 2019, 556, 320–329. [Google Scholar] [CrossRef]
  180. Gámez-Herrera, E.; García-Salinas, S.; Salido, S.; Sancho-Albero, M.; Andreu, V.; Pérez, M.; Luján, L.; Irusta, S.; Arruebo, M.; Mendoza, G. Drug-eluting wound dressings having sustained release of antimicrobial compounds. Eur. J. Pharm. Biopharm. 2020, 152, 327–339. [Google Scholar] [CrossRef]
  181. Kim, Y.G.; Lee, J.H.; Gwon, G.; Kim, S.I.; Park, J.G.; Lee, J. Essential Oils and Eugenols Inhibit Biofilm Formation and the Virulence of Escherichia coli O157:H7. Sci. Rep. 2016, 6, 36377. [Google Scholar] [CrossRef]
  182. Negut, I.; Grumezescu, V.; Ficai, A.; Grumezescu, A.M.; Holban, A.M.; Popescu, R.C.; Savu, D.; Vasile, B.S.; Socol, G. MAPLE deposition of nigella sativa functionalized Fe3O4 nanoparticles for antimicrobial coatings. Appl. Surf. Sci. 2018, 455, 513–521. [Google Scholar] [CrossRef]
  183. Ficai, D.; Grumezescu, V.; Fufă, O.M.; Popescu, R.C.; Holban, A.M.; Ficai, A.; Grumezescu, A.M.; Mogoanta, L.; Mogosanu, G.D.; Andronescu, E. Antibiofilm Coatings Based on PLGA and Nanostructured Cefepime-Functionalized Magnetite. Nanomaterials 2018, 8, 633. [Google Scholar] [CrossRef] [PubMed]
  184. Grumezescu, V.; Negut, I.; Grumezescu, A.M.; Ficai, A.; Dorcioman, G.; Socol, G.; Iordache, F.; Truşcă, R.; Vasile, B.S.; Holban, A.M. MAPLE fabricated coatings based on magnetite nanoparticles embedded into biopolymeric spheres resistant to microbial colonization. Appl. Surf. Sci. 2018, 448, 230–236. [Google Scholar] [CrossRef]
  185. Grumezescu, V.; Holban, A.M.; Grumezescu, A.M.; Socol, G.; Ficai, A.; Vasile, B.S.; Truscǎ, R.; Bleotu, C.; Lazar, V.; Chifiriuc, C.M.; et al. Usnic acid-loaded biocompatible magnetic PLGA-PVA microsphere thin films fabricated by maple with increased resistance to staphylococcal colonization. Biofabrication 2014, 6, 035002. [Google Scholar] [CrossRef] [PubMed]
  186. Zhang, Z.; Zhou, S.; Zhang, Y.; Wu, D.; Yang, X. The dual delivery of growth factors and antimicrobial peptide by PLGA/GO composite biofilms to promote skin-wound healing. New J. Chem. 2020, 44, 1463–1476. [Google Scholar] [CrossRef]
  187. Aniagyei, S.E.; Sims, L.B.; Malik, D.A.; Tyo, K.M.; Curry, K.C.; Kim, W.; Hodge, D.A.; Duan, J.; Steinbach-Rankins, J.M. Evaluation of poly(lactic-co-glycolic acid) and poly(dl-lactide-co-ε-caprolactone) electrospun fibers for the treatment of HSV-2 infection. Mater. Sci. Eng. C 2017, 72, 238–251. [Google Scholar] [CrossRef]
  188. Rădulescu, D.; Grumezescu, V.; Andronescu, E.; Holban, A.M.; Grumezescu, A.M.; Socol, G.; Oprea, A.E.; Rădulescu, M.; Surdu, A.; Trusca, R.; et al. Biocompatible cephalosporin-hydroxyapatite-poly(lactic-co-glycolic acid)-coatings fabricated by MAPLE technique for the prevention of bone implant associated infections. Appl. Surf. Sci. 2016, 374, 387–396. [Google Scholar] [CrossRef]
  189. Ghavimi, M.A.; Bani Shahabadi, A.; Jarolmasjed, S.; Memar, M.Y.; Maleki Dizaj, S.; Sharifi, S. Nanofibrous asymmetric collagen/curcumin membrane containing aspirin-loaded PLGA nanoparticles for guided bone regeneration. Sci. Rep. 2020, 10, 18200. [Google Scholar] [CrossRef]
  190. Song, W.; Xiao, Y. Sequential drug delivery of vancomycin and rhBMP-2 via pore-closed PLGA microparticles embedded photo-crosslinked chitosan hydrogel for enhanced osteointegration. Int. J. Biol. Macromol. 2021, 182, 612–625. [Google Scholar] [CrossRef]
  191. Eren Boncu, T.; Uskudar Guclu, A.; Catma, M.F.; Savaser, A.; Gokce, A.; Ozdemir, N. In vitro and in vivo evaluation of linezolid loaded electrospun PLGA and PLGA/PCL fiber mats for prophylaxis and treatment of MRSA induced prosthetic infections. Int. J. Pharm. 2020, 573, 118758. [Google Scholar] [CrossRef]
  192. Holešová, S.; Barabaszová, K.C.; Hundáková, M.; Ščuková, M.; Hrabovská, K.; Joszko, K.; Antonowicz, M.; Gzik-Zroska, B. Development of Novel Thin Polycaprolactone (PCL)/Clay Nanocomposite Films with Antimicrobial Activity Promoted by the Study of Mechanical, Thermal, and Surface Properties. Polymers 2021, 13, 3193. [Google Scholar] [CrossRef]
  193. Balcucho, J.; Narváez, D.M.; Castro-Mayorga, J.L. Antimicrobial and Biocompatible Polycaprolactone and Copper Oxide Nanoparticle Wound Dressings against Methicillin-Resistant Staphylococcus aureus. Nanomaterials 2020, 10, 1692. [Google Scholar] [CrossRef] [PubMed]
  194. Abdali, Z.; Logsetty, S.; Liu, S. Bacteria-Responsive Single and Core–Shell Nanofibrous Membranes Based on Polycaprolactone/Poly(ethylene succinate) for On-Demand Release of Biocides. ACS Omega 2019, 4, 4063–4070. [Google Scholar] [CrossRef] [PubMed]
  195. Lin, M.; Liu, Y.; Gao, J.; Wang, D.; Xia, D.; Liang, C.; Li, N.; Xu, R. Synergistic Effect of Co-Delivering Ciprofloxacin and Tetracycline Hydrochloride for Promoted Wound Healing by Utilizing Coaxial PCL/Gelatin Nanofiber Membrane. Int. J. Mol. Sci. 2022, 23, 1895. [Google Scholar] [CrossRef]
  196. Nasajpour, A.; Samandari, M.; Patil, C.D.; Abolhassani, R.; Suryawanshi, R.K.; Adelung, R.; Rubahn, H.-G.; Khademhosseini, A.; Mishra, Y.K.; Shukla, D.; et al. Nanoengineered Antiviral Fibrous Arrays with Rose-Thorn-Inspired Architectures. ACS Mater. Lett. 2021, 3, 1566–1571. [Google Scholar] [CrossRef]
  197. Figueroa-Lopez, K.J.; Vicente, A.A.; Reis, M.A.M.; Torres-Giner, S.; Lagaron, J.M. Antimicrobial and antioxidant performance of various essential oils and natural extracts and their incorporation into biowaste derived poly(3-hydroxybutyrate-co-3-hydroxyvalerate) layers made from electrospun ultrathin fibers. Nanomaterials 2019, 9, 144. [Google Scholar] [CrossRef] [PubMed]
  198. da Costa, R.C.; Daitx, T.S.; Mauler, R.S.; da Silva, N.M.; Miotto, M.; Crespo, J.S.; Carli, L.N. Poly(hydroxybutyrate-co-hydroxyvalerate)-based nanocomposites for antimicrobial active food packaging containing oregano essential oil. Food Packag. Shelf Life 2020, 26, 100602. [Google Scholar] [CrossRef]
  199. Figueroa-Lopez, K.J.; Enescu, D.; Torres-Giner, S.; Cabedo, L.; Cerqueira, M.A.; Pastrana, L.; Fuciños, P.; Lagaron, J.M. Development of electrospun active films of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) by the incorporation of cyclodextrin inclusion complexes containing oregano essential oil. Food Hydrocoll. 2020, 108, 106013. [Google Scholar] [CrossRef]
  200. Melendez-Rodriguez, B.; Figueroa-Lopez, K.J.; Bernardos, A.; Martínez-Máñez, R.; Cabedo, L.; Torres-Giner, S.; Lagaron, J.M. Electrospun antimicrobial films of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) containing eugenol essential oil encapsulated in mesoporous silica nanoparticles. Nanomaterials 2019, 9, 227. [Google Scholar] [CrossRef]
  201. Grumezescu, V.; Holban, A.M.; Iordache, F.; Socol, G.; Mogoşanu, G.D.; Grumezescu, A.M.; Ficai, A.; Vasile, B.Ş.; Truşcǎ, R.; Chifiriuc, M.C.; et al. MAPLE fabricated magnetite@eugenol and (3-hidroxybutyric acid-co-3-hidroxyvaleric acid)-polyvinyl alcohol microspheres coated surfaces with anti-microbial properties. Appl. Surf. Sci. 2014, 306, 16–22. [Google Scholar] [CrossRef]
  202. Grumezescu, V.; Holban, A.M.; Sima, L.E.; Chiritoiu, M.B.; Chiritoiu, G.N.; Grumezescu, A.M.; Ivan, L.; Safciuc, F.; Antohe, F.; Florica, C.; et al. Laser deposition of poly(3-hydroxybutyric acid-co-3-hydroxyvaleric acid)-lysozyme microspheres based coatings with anti-microbial properties. Int. J. Pharm. 2017, 521, 184–195. [Google Scholar] [CrossRef]
  203. Abdalkarim, S.Y.H.; Yu, H.Y.; Wang, D.; Yao, J. Electrospun poly(3-hydroxybutyrate-co-3-hydroxy-valerate)/cellulose reinforced nanofibrous membranes with ZnO nanocrystals for antibacterial wound dressings. Cellulose 2017, 24, 2925–2938. [Google Scholar] [CrossRef]
  204. Chen, Y.; Abdalkarim, S.Y.H.; Yu, H.Y.; Li, Y.; Xu, J.; Marek, J.; Yao, J.; Tam, K.C. Double stimuli-responsive cellulose nanocrystals reinforced electrospun PHBV composites membrane for intelligent drug release. Int. J. Biol. Macromol. 2020, 155, 330–339. [Google Scholar] [CrossRef]
  205. Hurtado, A.; Cano-Vicent, A.; Tuñón-Molin, A.; Aparicio-Collado, J.L.; Salesa, B.; Serra, R.S.; Serrano-Aroca, A. Engineering alginate hydrogel films with poly(3-hydroxybutyrate-co-3-valerate) and graphene nanoplatelets: Enhancement of antiviral activity, cell adhesion and electroactive properties. Int. J. Biol. Macromol. 2022, 219, 694–708. [Google Scholar] [CrossRef]
  206. Volova, T.G.; Golubev, A.I.; Nemtsev, I.V.; Lukyanenko, A.V.; Dudaev, A.E.; Shishatskaya, E.I. Laser Processing of Polymer Films Fabricated from PHAs Differing in Their Monomer Composition. Polymers 2021, 13, 1553. [Google Scholar] [CrossRef] [PubMed]
  207. Darie-Niță, R.N.; Râpă, M.; Frąckowiak, S. Special Features of Polyester-Based Materials for Medical Applications. Polymers 2022, 14, 951. [Google Scholar] [CrossRef] [PubMed]
  208. Serrano-Aroca, Á.; Cano-Vicent, A.; Sabater i Serra, R.; El-Tanani, M.; Aljabali, A.A.A.; Tambuwala, M.M.; Mishra, Y.K. Scaffolds in the microbial resistant era: Fabrication, materials, properties and tissue engineering applications. Mater. Today Bio. 2022, 16, 100412. [Google Scholar] [CrossRef]
  209. Parham, S.; Kharazi, A.Z.; Bakhsheshi-Rad, H.R.; Kharaziha, M.; Ismail, A.F.; Sharif, S.; Razzaghi, M.; Krishna, S.R.; Berto, F. Antimicrobial Synthetic and Natural Polymeric Nanofibers as Wound Dressing: A Review. Adv. Eng. Mater. 2022, 2101460. [Google Scholar] [CrossRef]
  210. Arif, Z.U.; Khalid, M.Y.; Noroozi, R.; Sadeghianmaryan, A.; Jalalvand, M.; Hossaine, M. Recent advances in 3D-printed polylactide and polycaprolactone-based biomaterials for tissue engineering applications. Int. J. Biol. Macromol. 2022, 218, 930–968. [Google Scholar] [CrossRef]
  211. Abbasian, M.; Massoumi, B.; Mohammad-Rezaei, R.; Samadian, H.; Jaymand, M. Scaffolding polymeric biomaterials: Are naturally occurring biological macromolecules more appropriate for tissue engineering? Int. J. Biol. Macromol. 2019, 134, 673–694. [Google Scholar] [CrossRef]
  212. Reddy, M.S.B.; Ponnamma, D.; Choudhary, R.; Sadasivuni, K.K. A Comparative Review of Natural and Synthetic Biopolymer Composite Scaffolds. Polymers 2021, 13, 1105. [Google Scholar] [CrossRef]
  213. Lian, X.; Liu, H.; Wang, X.; Xu, S.; Cui, F.; Bai, X. Antibacterial and biocompatible properties of vancomycin-loaded nano-hydroxyapatite/collagen/poly (lactic acid) bone substitute. Prog. Nat. Sci. Mater. Int. 2013, 23, 549–556. [Google Scholar] [CrossRef]
  214. Sadaba, N.; Larrañaga, A.; Orpella-Aceret, G.; Bettencourt, A.F.; Martin, V.; Biggs, M.; Ribeiro, I.A.; Ugartemendia, J.M.; Sarasua, J.-R.; Zuza, E. Benefits of Polydopamine as Particle/Matrix Interface in Polylactide/PD-BaSO4 Scaffolds. Int. J. Mol. Sci. 2020, 21, 5480. [Google Scholar] [CrossRef]
  215. Preis, E.; Anders, T.; Širc, J.; Hobzova, R.; Cocarta, A.-I.; Bakowsky, U.; Jedelská, J. Biocompatible indocyanine green loaded PLA nanofibers for in situ antimicrobial photodynamic therapy. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 115, 111068. [Google Scholar] [CrossRef] [PubMed]
  216. Jiao, J.; Peng, C.; Li, C.; Qi, Z.; Zhan, J.; Pan, S. Dual bio-active factors with adhesion function modified electrospun fibrous scaffold for skin wound and infections therapeutics. Sci. Rep. 2021, 11, 457. [Google Scholar] [CrossRef] [PubMed]
  217. Liu, X.; Nielsen, L.H.; Kłodzińska, S.N.; Nielsen, H.M.; Qu, H.; Christensen, L.P.; Rantanen, J.; Yang, M. Ciprofloxacin-loaded sodium alginate/poly (lactic-co-glycolic acid) electrospun fibrous mats for wound healing. Eur. J. Pharm. Biopharm. 2018, 123, 42–49. [Google Scholar] [CrossRef]
  218. Choipang, C.; Chuysinuan, P.; Suwantong, O.; Ekabutr, P.; Supaphol, P. Hydrogel wound dressings loaded with PLGA/ciprofloxacin hydrochloride nanoparticles for use on pressure ulcers. J. Drug Deliv. Sci. Technol. 2018, 47, 106–114. [Google Scholar] [CrossRef]
  219. Govoni, M.; Lamparelli, E.P.; Ciardulli, M.C.; Santoro, A.; Oliviero, A.; Palazzo, I.; Reverchon, E.; Vivarelli, L.; Maso, A.; Storni, E.; et al. Demineralized bone matrix paste formulated with biomimetic PLGA microcarriers for the vancomycin hydrochloride controlled delivery: Release profile, citotoxicity and efficacy against S. aureus. Int. J. Pharm. 2020, 582, 119322. [Google Scholar] [CrossRef]
  220. Ajmal, G.; Bonde, G.V.; Mittal, P.; Khan, G.; Pandey, V.K.; Bakade, B.V.; Mishra, B. Biomimetic PCL-gelatin based nanofibers loaded with ciprofloxacin hydrochloride and quercetin: A potential antibacterial and anti-oxidant dressing material for accelerated healing of a full thickness wound. Int. J. Pharm. 2019, 567, 118480. [Google Scholar] [CrossRef]
  221. Jafari, A.; Amirsadeghi, A.; Hassanajili, S.; Azarpira, N. Bioactive antibacterial bilayer PCL/gelatin nanofibrous scaffold promotes full-thickness wound healing. Int. J. Pharm. 2020, 583, 119413. [Google Scholar] [CrossRef]
  222. Baghersad, S.; Hivechi, A.; Bahrami, S.H.; Milan, P.B.; Siegel, R.A.; Amoupour, M. Optimal Aloe vera encapsulated PCL/Gel nanofiber design for skin substitute application and the evaluation of its in vivo implantation. J. Drug Deliv. Sci. Technol. 2022, 74, 103536. [Google Scholar] [CrossRef]
  223. Asghari, F.; Faradonbeh, D.R.; Malekshahi, Z.V.; Nekounam, H.; Ghaemi, B.; Yousefpoor, Y.; Ghanbari, H.; Faridi-Majidi, R. Hybrid PCL/chitosan-PEO nanofibrous scaffolds incorporated with A. euchroma extract for skin tissue engineering application. Carbohydr. Polym. 2022, 278, 118926. [Google Scholar] [CrossRef] [PubMed]
  224. Ayran, M.; Dirican, A.Y.; Saatcioglu, E.; Ulag, S.; Sahin, A.; Aksu, B.; Croitoru, A.-M.; Ficai, D.; Gunduz, O.; Ficai, A. 3D-Printed PCL Scaffolds Combined with Juglone for Skin Tissue Engineering. Bioengineering 2022, 9, 427. [Google Scholar] [CrossRef] [PubMed]
  225. Budi, H.S.; Ansari, M.J.; Jasim, S.A.; Abdelbasset, W.K.; Bokov, D.; Mustafa, Y.F.; Najm, M.A.A.; Kazemnejadi, M. Preparation of antibacterial Gel/PCL nanofibers reinforced by dicalcium phosphate-modified graphene oxide with control release of clindamycin for possible application in bone tissue engineering. Inorg. Chem. Commun. 2022, 139, 109336. [Google Scholar] [CrossRef]
  226. de Almeida Neto, G.R.; Barcelos, M.V.; Ribeiro, M.E.A.; Folly, M.M.; Rodríguez, R.J.S. Formulation and characterization of a novel PHBV nanocomposite for bone defect filling and infection treatment. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 104, 110004. [Google Scholar] [CrossRef]
  227. Dalgic, A.D.; Koman, E.; Karatas, A.; Tezcaner, A.; Keskin, D. Natural origin bilayer pullulan-PHBV scaffold for wound healing applications. Biomater. Adv. 2022, 134, 112554. [Google Scholar] [CrossRef]
  228. Kamal, R.; Razzaq, A.; Ali shah, K.; Khan, Z.U.; Khan, N.U.; Menaa, F.; Iqbal, H.; Cui, J. Evaluation of cephalexin-loaded PHBV nanofibers for MRSA-infected diabetic foot ulcers treatment. J. Drug Deliv. Sci. Technol. 2022, 71, 103349. [Google Scholar] [CrossRef]
  229. Ahmed, T.A.; Aljaeid, B.M. A potential in situ gel formulation loaded with novel fabricated poly(lactide-co-glycolide) nanoparticles for enhancing and sustaining the ophthalmic delivery of ketoconazole. Int. J. Nanomed. 2017, 12, 1863–1875. [Google Scholar] [CrossRef]
  230. Gebreel, R.M.; Edris, N.A.; Elmofty, H.M.; Tadros, M.I.; El-Nabarawi, M.A.; Hassan, D.H. Development and Characterization of PLGA Nanoparticle-Laden Hydrogels for Sustained Ocular Delivery of Norfloxacin in the Treatment of Pseudomonas Keratitis: An Experimental Study. Drug Des. Dev. Ther. 2020, 15, 399–418. [Google Scholar] [CrossRef]
  231. Polat, H.K.; Pehlivan, S.B.; Özkul, C.; Çalamak, S.; Öztürk, N.; Aytekin, E.; Fırat, A.; Ulubayram, K.; Kocabeyoğlu, S.; İrkeç, M.; et al. Development of besifloxacin HCl loaded nanofibrous ocular inserts for the treatment of bacterial keratitis: In vitro, ex vivo and in vivo evaluation. Int. J. Pharm. 2020, 585, 119552. [Google Scholar] [CrossRef]
  232. Yousry, C.; Elkheshen, S.A.; El-laithy, H.M.; Essam, T.; Fahmy, R.H. Studying the influence of formulation and process variables on Vancomycin-loaded polymeric nanoparticles as potential carrier for enhanced ophthalmic delivery. Eur. J. Pharm. Sci. 2017, 100, 142–154. [Google Scholar] [CrossRef]
  233. Cheng, Y.-H.; Chang, Y.-F.; Ko, Y.-C.; Liu, C.J. Development of a dual delivery of levofloxacin and prednisolone acetate via PLGA nanoparticles/ thermosensitive chitosan-based hydrogel for postoperative management: An in-vitro and ex-vivo study. Int. J. Biol. Macromol. 2021, 180, 365–374. [Google Scholar] [CrossRef] [PubMed]
  234. Sun, M.; Zhu, C.; Long, J.; Lu, C.; Pan, X.; Wu, C. PLGA microsphere-based composite hydrogel for dual delivery of ciprofloxacin and ginsenoside Rh2 to treat Staphylococcus aureus-induced skin infections. Drug Deliv. 2020, 27, 632–641. [Google Scholar] [CrossRef] [PubMed]
  235. Pettinelli, N.; Rodríguez-Llamazares, S.; Farrag, Y.; Bouza, R.; Barral, L.; Feijoo-Bandín, S.; Lago, F. Poly(hydroxybutyrate-co-hydroxyvalerate) microparticles embedded in κ-carrageenan/locust bean gum hydrogel as a dual drug delivery carrier. Int. J. Biol. Macromol. 2020, 146, 110–118. [Google Scholar] [CrossRef]
  236. Mendes, A.I.; Rebelo, R.; Aroso, I.; Correlo, V.M.; Fraga, A.G.; Pedrosa, J.; Marques, A.P. Development of an antibiotics delivery system for topical treatment of the neglected tropical disease Buruli ulcer. Int. J. Pharm. 2022, 623, 121954. [Google Scholar] [CrossRef] [PubMed]
  237. Zhang, S.; Chen, G.; Wang, M.; Lin, B.; Gao, X.; Hu, J.; Chen, B.; Zhang, C. Osteogenic and anti-inflammatory potential of oligochitosan nanoparticles in treating osteomyelitis. Biomater. Adv. 2022, 135, 112681. [Google Scholar] [CrossRef] [PubMed]
  238. Yuan, B.; Zhang, Y.; Wang, Q.; Ren, G.; Wang, Y.; Zhou, S.; Wang, Q.; Peng, C.; Cheng, X. Thermosensitive vancomycin@PLGA-PEG-PLGA/HA hydrogel as an all-in-one treatment for osteomyelitis. Int. J. Pharm. 2022, 627, 122225. [Google Scholar] [CrossRef]
  239. Lee, J.-H.; Park, J.-K.; Son, K.-H.; Lee, J.-W. PCL/Sodium-Alginate Based 3D-Printed Dual Drug Delivery System with Antibacterial Activity for Osteomyelitis Therapy. Gels 2022, 8, 163. [Google Scholar] [CrossRef]
  240. Motealleh, A.; Kehr, N.S. Nanocomposite Hydrogels and Their Applications in Tissue Engineering. Adv. Healthcare Mater. 2017, 6, 1600938. [Google Scholar] [CrossRef]
  241. Choi, A.H.; Ben-Nissan, B.; Matinlinna, J.P.; Conway, R.C.J. Current Perspectives: Calcium Phosphate Nanocoatings and Nanocomposite Coatings in Dentistry. Dent. Res. 2013, 92, 853. [Google Scholar] [CrossRef]
  242. Spizzirri, U.G.; Curcio, M.; Cirillo, G.; Spataro, T.; Vittorio, O.; Picci, N.; Hampel, S.; Iemma, F.; Nicoletta, F.P. Recent Advances in the Synthesis and Biomedical Applications of Nanocomposite Hydrogels. Pharmaceutics 2015, 7, 413. [Google Scholar] [CrossRef] [Green Version]
  243. Sensenig, R.; Sapir, Y.; MacDonald, C.; Cohen, S.; Polyak, B. Magnetic nanoparticle-based approaches to locally target therapy and enhance tissue regeneration in vivo. Nanomedicine 2012, 7, 1425. [Google Scholar] [CrossRef] [PubMed]
  244. Wang, X.; Law, J.; Luo, M.; Gong, Z.; Yu, J.; Tang, W.; Zhang, Z.; Mei, X.; Huang, Z.; You, L.; et al. Magnetic Measurement and Stimulation of Cellular and Intracellular Structures. ACS Nano 2020, 14, 3805. [Google Scholar] [CrossRef] [PubMed]
  245. Colombo, M.; Carregal-Romero, S.; Casula, M.F.; Gutiérrez, L.; Morales, M.P.; Böhm, I.B.; Heverhagen, J.T.; Prosperi, D.; Parak, W.J. Biological applications of magnetic nanoparticles. Chem. Soc. Rev. 2012, 41, 4306. [Google Scholar] [CrossRef] [PubMed]
  246. Berman, S.M.C.; Walczak, P.; Bulte, J.W. Tracking stem cells using magnetic nanoparticles. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2011, 3, 343–355. [Google Scholar] [CrossRef]
  247. Bonnemay, L.; Hoffmann, C.; Gueroui, Z. Remote control of signaling pathways using magnetic nanoparticles. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2015, 7, 342–354. [Google Scholar] [CrossRef]
  248. Henstock, J.R.; Rotherham, M.; El Haj, A.J. Magnetic ion channel activation of TREK1 in human mesenchymal stem cells using nanoparticles promotes osteogenesis in surrounding cells. J. Tissue Eng. 2018, 9, 204173141880869. [Google Scholar] [CrossRef] [PubMed]
  249. Rotherham, M.; Nahar, T.; Goodman, T.; Telling, N.; Gates, M.; El Haj, A. Magnetic Mechanoactivation of Wnt Signaling Augments Dopaminergic Differentiation of Neuronal Cells. Adv. Biosyst. 2019, 3, 1900091. [Google Scholar] [CrossRef]
  250. Sapir, Y.; Cohen, S.; Friedman, G.; Polyak, B. The promotion of in vitro vessel-like organization of endothelial cells in magnetically responsive alginate scaffolds. Biomaterials 2012, 33, 4100. [Google Scholar] [CrossRef]
  251. Sapir, Y.; Polyak, B.; Cohen, S. Cardiac Tissue Engineering in Magnetically Actuated Scaffolds. Nanotechnology 2014, 25, 014009. [Google Scholar] [CrossRef]
  252. Peng, J.; Zhao, J.; Long, Y.; Xie, Y.; Nie, J.; Chen, L. Magnetic Materials in Promoting Bone Regeneration. Front. Mater. Sec. Biomater. 2019, 6, 268. [Google Scholar] [CrossRef]
  253. Sapir, Y.; Ruvinov, E.; Polyak, B.; Cohen, S. Magnetically Actuated Alginate Scaffold: A Novel Platform for Promoting Tissue Organization and Vascularization. In Cardiac Tissue Engineering. Methods in Molecular Biology; Radisic, M., Black, L., III, Eds.; Humana Press: New York, NY, USA, 2014; Volume 1181. [Google Scholar] [CrossRef]
  254. Tognato, R.; Armiento, A.R.; Bonfrate, V.; Levato, R.; Malda, J.; Alini, M.; Eglin, D.; Giancane, G.; Serra, T. A Stimuli-Responsive Nanocomposite for 3D Anisotropic Cell-Guidance and Magnetic Soft Robotics. Adv. Funct. Mater. 2019, 29, 1804647. [Google Scholar] [CrossRef]
  255. Esmaeili, E.; Soleimani, M.; Ghiass, M.A.; Hatamie, S.; Vakilian, S.; Zomorrod, M.S. Magnetoelectric nanocomposite scaffold for high yield differentiation of mesenchymal stem cells to neural-like cells. J. Cell Physiol. 2019, 234, 13617–13628. [Google Scholar] [CrossRef] [PubMed]
  256. Discher, D.E.; Mooney, D.J.; Zandstra, P.W. Growth factors, matrices, and forces combine and control stem cells. Science 2009, 324, 1673–1677. [Google Scholar] [CrossRef] [PubMed]
  257. Li, W.; Jiang, K.; Ding, S. Concise review: A chemical approach to control cell fate and function. Stem Cells 2012, 30, 61–68. [Google Scholar] [CrossRef]
  258. Liu, S.; Takahashi, M.; Kiyoi, T.; Toyama, K.; Mogi, M. Genetic Manipulation of Calcium Release-Activated Calcium Channel 1 Modulates the Multipotency of Human Cartilage-Derived Mesenchymal Stem Cells. J. Immunol. Res. 2019, 2019, 7510214. [Google Scholar] [CrossRef]
  259. Argani, H. Anti-HLA Antibody: The Role of Epitopes in Organ Transplantation. Exp. Clin. Transplant. 2019, 17 (Suppl. S1), 38–42. [Google Scholar] [CrossRef]
  260. Takata, N.; Eiraku, M. Stem cells and genome editing: Approaches to tissue regeneration and regenerative medicine. J. Hum. Genet. 2018, 63, 165–178. [Google Scholar] [CrossRef]
  261. Huebsch, N.; Arany, P.R.; Mao, A.S.; Shvartsman, D.; Ali, O.A.; Bencherif, S.A.; Rivera-Feliciano, J.; Mooney, D.J. Harnessing traction-mediated manipulation of the cell/matrix interface to control stem-cell fate. Nat. Mater. 2010, 9, 518. [Google Scholar] [CrossRef]
  262. Moghaddam, M.M.; Bonakdar, S.; Shokrgozar, M.A.; Zaminy, A.; Vali, H.; Faghihi, S. Engineered substrates with imprinted cell-like topographies induce direct differentiation of adipose-derived mesenchymal stem cells into Schwann cells. Artif. Cells Nanomed. Biotechnol. 2019, 47, 1022–1035. [Google Scholar] [CrossRef]
  263. Fu, C.Y.; Lin, C.Y.; Chu, W.C.; Chang, H.Y. A simple cell patterning method using magnetic particle-containing photosensitive poly (ethylene glycol) hydrogel blocks: A technical note. Tissue Eng. Part C 2011, 17, 871–877. [Google Scholar] [CrossRef]
  264. Wu, S.; Yu, Q.; Sun, Y.; Tian, J. Synergistic effect of a LPEMF and SPIONs on BMMSC proliferation, directional migration, and osteoblastogenesis. Am. J. Transl. Res. 2018, 10, 1431–1443. [Google Scholar] [PubMed]
  265. Huang, D.M.; Hsiao, J.K.; Chen, Y.C.; Chien, L.Y.; Yao, M.; Chen, Y.K.; Ko, B.S.; Hsu, S.C.; Tai, L.A.; Cheng, H.Y.; et al. The promotion of human mesenchymal stem cell proliferation by uperparamagnetic iron oxide nanoparticles. Biomaterials 2009, 30, 3645–3651. [Google Scholar] [CrossRef]
  266. Daňková, J.; Buzgo, M.; Vejpravová, J.; Kubíčková, S.; Sovková, V.; Vysloužilová, L.; Mantlíková, A.; Nečas, A.; Amler, E. Highly efficient mesenchymal stem cell proliferation on poly-ε-caprolactone nanofibers with embedded magnetic nanoparticles. Int. J. Nanomed. 2015, 10, 7307. [Google Scholar] [CrossRef]
  267. Wang, Q.; Chen, B.; Cao, M.; Sun, J.; Wu, H.; Zhao, P.; Xing, J.; Yang, Y.; Zhang, X.; Ji, M.; et al. Biomedical polymers: Synthesis, properties, and applications. Biomaterials 2016, 86, 11. [Google Scholar] [CrossRef] [PubMed]
  268. Nakamichi, N.; Ishioka, Y.; Hirai, T.; Ozawa, S.; Tachibana, M.; Nakamura, N.; Takarada, T.; Yoneda, Y. Possible promotion of neuronal differentiation in fetal rat brain neural progenitor cells after sustained exposure to static magnetism. J. Neurosci. Res. 2009, 87, 2406. [Google Scholar] [CrossRef]
  269. Miyakoshi, J. Effects of static magnetic fields at the cellular level. Prog. Biophys. Mol. Biol. 2005, 87, 213. [Google Scholar] [CrossRef] [PubMed]
  270. Marycz, K.; Kornicka, K.; Röcken, M. Static Magnetic Field (SMF) as a Regulator of Stem Cell Fate—New Perspectives in Regenerative Medicine Arising from an Underestimated Tool. Stem Cell Rev. Rep. 2018, 14, 785–792. [Google Scholar] [CrossRef]
  271. Pardo, A.; Gómez-Florit, M.; Barbosa, S.; Taboada, P.; Domingues, R.M.A.; Gomes, M.E. Magnetic Nanocomposite Hydrogels for Tissue Engineering: Design Concepts and Remote Actuation Strategies to Control Cell Fate. ACS Nano 2021, 15, 175–209. [Google Scholar] [CrossRef]
  272. Fernández-Arias, M.; Vilas, A.M.; Boutinguiza, M.; Rodríguez, D.; Arias-González, F.; Pou-Álvarez, P.; Riveiro, A.; Gil, J.; Pou, J. Palladium Nanoparticles Synthesized by Laser Ablation in Liquids for Antimicrobial Applications. Nanomaterials 2022, 12, 2621. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of antimicrobial biopolyester-based formulations.
Figure 1. Schematic representation of antimicrobial biopolyester-based formulations.
Ijms 24 02945 g001
Figure 2. Schematic representation of degradation mechanisms in biopolyester-based formulations.
Figure 2. Schematic representation of degradation mechanisms in biopolyester-based formulations.
Ijms 24 02945 g002
Figure 3. Schematic representation of synthesis methods used to fabricate biopolyester-based layered formulations.
Figure 3. Schematic representation of synthesis methods used to fabricate biopolyester-based layered formulations.
Ijms 24 02945 g003
Table 1. Particulate biocompatible polyester-based antiviral formulations.
Table 1. Particulate biocompatible polyester-based antiviral formulations.
System DescriptionTherapeutic PotentialRefs.
PLA oligomersInhibitory and virucidal effects against human Influenza A virus (IAV) and severe acute respiratory syndrome (SARS) virus[128]
PLA/Ag nanocomposites (green tea extract reduced metallic particles)Virucidal effects against human IAV and adenovirus serotype 2 [129]
PLA nanoparticles entrapping bacterial antigen adjuvanted with aluminum hydroxideLong-lasting and efficient protection against infection caused by SARS coronavirus 2 (SARS-CoV-2)[130]
Curcumin-loaded PLGA nanoparticlesControlled and targeted local management of SARS-CoV-2 infection through antimicrobial photodynamic therapy [131]
Oseltamivir-loaded PEGylated PLGA nanoparticlesEffective targeted treatment of pulmonary cancer and IAV infection[132]
Peptide-conjugated maleimine-functionalized PLGA nanoparticles encapsulating lamivudineControlled and targeted local treatment of hepatitis B virus (HBV) infection[133]
Adefovir-loaded PLGA microspheres[134]
PLGA-CS microparticles encapsulating interferon-alphaEffective protection against mengovirus infection[135]
PLGA nanoparticles entrapping protein or peptide antigens and immunostimulatory adjuvantsEffective protection and targeted local treatment of IAV infection[136]
PLGA nanoparticles entrapping viral antigen adjuvanted with pattern-recognition receptor agonists[137]
PLGA microparticles loaded with viral nucleoprotein adjuvanted with immunostimulatory agonists and carbomer–lecithin nanoemulsion Effective protection and targeted local treatment of IAV and SARS-CoV-2 infection[138]
Nanoparticles of PLGA-PEG and PCL grafted with membrane receptor ligands and loaded with remdesivirEfficient and targeted local treatment of SARS-CoV-2 infection[139]
Blank PCL nanocapsules with Eudragit surface coatingSelective inhibitory effects against Herpes simplex virus (HSV) type-1[140]
Cidofovir-loaded PEG-PCL nanoparticles ink formulationControlled and prolonged efficiency for the local treatment of human papilloma virus (HPV) infection [141,142]
Table 2. Biopolyester-based gel formulations with antimicrobial activity.
Table 2. Biopolyester-based gel formulations with antimicrobial activity.
System DescriptionTherapeutic EffectsRefs.
Ketoconazole-entrapped PLGA nanoparticles loaded into alginate-chitosan in situ gel formulations Augmented drug permeation and sustained drug release
Treatment of Candida albicans fungal keratitis and endophthalmitis
[229]
Norfloxacin-loaded PLGA nanoparticles incorporated within hydroxypropyl methylcellulose hydrogels Prolonged drug release and superior biosafety profile
Treatment of P. aeruginosa keratitis
[230]
Besifloxacin-loaded PCL/PEG nanofibrous inserts entrapped within thiolated sodium alginate[231]
Vancomycin-loaded PLGA/Eudragit and PCL/Eudragit nanoparticles incorporated in Carbopol-based hydrogelsProlonged drug release and superior biosafety profile
Treatment of S. aureus keratitis
[232]
Levofloxacin-encapsulated PLGA nanoparticles embedded in prednisolone-containing CS/gelatin hydrogelsExtended dual-drug release and improved ocular bioavailability
Treatment of S. aureus keratitis, management of endophthalmitis
[233]
Ciprofloxacin-encapsulated PLGA microspheres loaded within poloxamer/hyaluronic acid hydrogels entrapping ginsenosideSequential release ability (short-term release of the immunomodulatory ginsenoside and long-term release of the antibiotic)
Treatment of skin infections through synergistic efficiency against methicillin-sensitive and methicillin-resistant S. aureus
[234]
Mupirocin-/ketoprofen-co-encapsulated mesoporous PHBV microparticles embedded in κ-carrageenan/locust bean gum hydrogelsThermo-sensitive and prolonged dual-drug release
Potential wound-healing applications
[235]
Rifampicin-loaded PHBV microparticles embedded in streptomycin-containing gellan gum hydrogelsSustained dual-drug release
Treatment of skin ulcers caused by Mycobacterium ulcerans infection
[236]
Vancomycin-loaded oligochitosan nanoparticles mixed with PLGA-PEG-PLGA gelsThermo-sensitive and sustained drug release, osteogenic differentiation ability, and important antibacterial and anti-biofilm effects against S. aureus and S. aureus mutans, respectively
Treatment of osteomyelitis and regeneration of infected bone tissue
[237]
Vancomycin-embedded and HAp-loaded PLGA-PEG-PLGA gels[238]
Cefazoline-loaded PCL scaffold encapsulated in rifampicin-containing alginate hydrogelsProlonged dual-drug release and important antibacterial and anti-biofilm effects against S. aureus
Treatment of osteomyelitis
[239]
Osteogenic protein-entrapped PLGA microspheres loaded in vancomycin-containing CS hydrogelsSequential release ability (fast release of the antibiotic for 2 days and sustained long-term release of the protein for 12 days)
Important antibacterial activity and reduced infection-mediated inflammation caused by S. aureus mutans, osteogenic differentiation, and bone regeneration ability
[191]
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

Gherasim, O.; Grumezescu, V.; Irimiciuc, S.A. Overview of Antimicrobial Biodegradable Polyester-Based Formulations. Int. J. Mol. Sci. 2023, 24, 2945. https://doi.org/10.3390/ijms24032945

AMA Style

Gherasim O, Grumezescu V, Irimiciuc SA. Overview of Antimicrobial Biodegradable Polyester-Based Formulations. International Journal of Molecular Sciences. 2023; 24(3):2945. https://doi.org/10.3390/ijms24032945

Chicago/Turabian Style

Gherasim, Oana, Valentina Grumezescu, and Stefan Andrei Irimiciuc. 2023. "Overview of Antimicrobial Biodegradable Polyester-Based Formulations" International Journal of Molecular Sciences 24, no. 3: 2945. https://doi.org/10.3390/ijms24032945

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