Next Article in Journal
Optimize PLA/EVA Polymers Blend Compositional Coating for Next Generation Biodegradable Drug-Eluting Stents
Next Article in Special Issue
Influence of Aging on the Fracture Characteristics of Polyetheretherketone Dental Crowns: A Preliminary Study
Previous Article in Journal
Obtaining and Characterizing Composite Biomaterials of Animal Resources with Potential Applications in Regenerative Medicine
Previous Article in Special Issue
A Tumor Accelerator Based on Multicomponent Bone Scaffolds and Cancer Cell Homing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Insights on Development Aspects of Polymeric Nanocarriers: The Translation from Bench to Clinic

1
M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, Haryana, India
2
Formulation Research and Development, Mankind Research Centre, Manesar, Gurugram 122050, Haryana, India
3
Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Saifai, Etawah 206130, Uttar Pradesh, India
4
Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, Haryana, India
5
Department of Pharmacy, G.D. Goenka University, Sohna Road, Gurugram 122103, Haryana, India
6
Biology Department, Faculty of Science and Arts in Dahran Aljnoub, King Khalid University, Abha 62529, Saudi Arabia
7
Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 61413, Saudi Arabia
8
Agriculture Research Centre, Soil, Water and Environment Research Institute, Giza 3725004, Egypt
9
Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea
10
Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
*
Authors to whom correspondence should be addressed.
Polymers 2022, 14(17), 3545; https://doi.org/10.3390/polym14173545
Submission received: 14 June 2022 / Revised: 24 August 2022 / Accepted: 25 August 2022 / Published: 29 August 2022

Abstract

:
Scientists are focusing immense attention on polymeric nanocarriers as a prominent delivery vehicle for several biomedical applications including diagnosis of diseases, delivery of therapeutic agents, peptides, proteins, genes, siRNA, and vaccines due to their exciting physicochemical characteristics which circumvent degradation of unstable drugs, reduce toxic side effects through controlled release, and improve bioavailability. Polymers-based nanocarriers offer numerous benefits for in vivo drug delivery such as biocompatibility, biodegradability, non-immunogenicity, active drug targeting via surface modification, and controlled release due to their pH—and thermosensitive characteristics. Despite their potential for medicinal use, regulatory approval has been achieved for just a few. In this review, we discuss the historical development of polymers starting from their initial design to their evolution as nanocarriers for therapeutic delivery of drugs, peptides, and genes. The review article also expresses the applications of polymeric nanocarriers in the pharmaceutical and medical industry with a special emphasis on oral, ocular, parenteral, and topical application of drugs, peptides, and genes over the last two decades. The review further examines the practical, regulatory, and clinical considerations of the polymeric nanocarriers, their safety issues, and directinos for future research.

Graphical Abstract

1. Introduction

A polymer represents several repeating units leading to the formation of a compound that has characteristics of high relative molecular mass and associated properties. In 1833, Jons Jakob Berzelius coined the term “polymer” which originated from the Greek word (polus, meaning “many, much”) and (meros, meaning “part”). Following major developments in polymer fabrication, the molecular properties of polymers were only recognized after the efforts of Hermann Staudinger in 1922, who first suggested that polymers consist of long chains of atoms bound together by covalent bonds. This took more than a decade to achieve acceptance among the scientific community. Initially, polymers were investigated according to the theory of associations or composite theory that emerged in 1861. It was suggested that cellulose and other polymers were colloids and molecular aggregates with small molecular weights bound by an unidentified intermolecular force. The use of polymers is not new in the medical field [1].
Natural polymers are being used for decades as ingredients for herbal remedies. The case is very different when it comes to synthetic polymers. Water-soluble polymers can be considered a modern accomplishment as macromolecular drugs or as part of the inoculation-related drug delivery systems. The first polymer-drug conjugate in form of mescaline-N-vinylpyrrolidone conjugates was synthesized in 1955. A decade later, it was predicted that polyethylene glycol (PEG) can be attached to proteins using a method called PEGylation that comprises adding the covalent bond of polymer (ethylene glycol) polymer chains to some other entity, normally a therapeutically active drug or protein. In 1994, the first synthetic polymer-drug conjugate was clinically proven to treat cancer. This was made up of a doxorubicin copolymer conjugation of (N-(2-hydroxypropyl) methacrylamide [2]. In 1977, Davis and Abuchowski described first-time PEGylation for drug delivery via the covalent attachment of PEG to bovine serum albumin and liver catalase proteins [3]. They reported that it could increase the systemic circulation time and decrease the immunogenicity of the proteins without significantly compromising activity. In 1990, the FDA approved the first PEGylated protein product, Adagen®, a PEGylated adenosine deaminase enzyme for severe combined immunodeficiency disease. Doxil® is the first FDA-approved PEGylated nanoparticle (NP) based product that was commercialized in 1995. At present, eight other PEGylated protein therapeutics have been FDA approved for various diseases [4]. The success of protein PEGylation as a method for producing longer circulating, and thus more efficacious intravenous therapies, led to investigations of nanoparticle (NP) PEGylation for systemic applications in the early 1980s and 1990s [5]. Recognized as foreign objects, they are readily excreted from the systemic circulation by the cells of the mononuclear phagocyte system, precluding accumulation in target cells and tissues [6,7,8]. Block copolymer-based micelles have also been explored for targeting specific cancer cells as these micelles are capable of trapping the drug or connecting it covalently to the co-polymers. Two polymer-protein complexes, PEG-interferon-α (an antiviral medication meant to treat liver cirrhosis) and PEG granulocyte colony-stimulating factor were put on the market around the 2000s [9]. The first therapeutic nanoparticles (albumin-trapped paclitaxel) were licensed for metastatic breast cancer treatment after five years [10]. Since then, polymeric micelles and nanoparticles comprising block copolymers have emerged as a storm and impacted clinical care. Some of the polymeric micelles have been approved and are currently being marketed in Korea and Europe. Genexol-PM is a polymeric micellar formulation of paclitaxel got approved in 2007 and is marketed under the name Cynviloq by Sorrento Pharmaceutics [11]. In 2013, NanoCarrier Co. Ltd. Tokyocompleted Phase II clinical studies of Nanoplatin (NC-6004) which is a polymeric micellar formulation of cisplatin [12]. NK012 of Nippon Kayaku Co. Ltd. Tokyo, is another polymeric micelle formulation of SN-38 (an irinotecan metabolite) which has completed Phase II clinical trials in 2015 and received orphan drug designation from USFDA [13]. In 2017, Nippon Kayaku Co., Ltd., Tokyo completed another clinical Phase III study for NK105 which is a polymeric micellar formulation of paclitaxel consisting of PEG and modified polyaspartate as a hydrophobic block. Currently, Docetaxel-loaded polymeric micelles plus Oxaliplatin is under Phase II clinical trial for patients with esophageal squamous cell carcinoma. Epirubicin-loaded polymeric nanoparticles is undergoing a Phase I/II clinical trial for patients having tumors or soft tissue sarcoma. Another polymeric nanoparticle loaded with Cetuximab is undergoing a Phase I clinical trial for colon cancer [14]. All of the above accomplishments represent the central factor that led to the growth of polymer-based pharmaceutical products, viz. nanoparticles, nanoconjugates, and micelles.
Nanomaterials took the pharmaceutical, nutritional supplements, and food industries by storm. Such structures are 1–100 nm (10−9 m) in diameter and have distinct physicochemical properties in comparison to macromaterials [15]. Nanotechnology is one of the most innovative technologies and is capable of improving food safety and quality. However, there are many studies showings that nanomaterials are not inherently harmless [16]. Nanomaterials are used for therapeutics in the healthcare sector, treatment for cancer, and diagnosis of disease. But in addition to their advantages, they also possess several undesirable effects, among which an increase in the absorption rate is the most important [17,18,19]. They increase the absorption rate as a result of the increased surface area of the nanomaterials, which allows increased absorption rate by the surrounding tissues leading to the generation of several unintended results [20]. The increased absorption rate is often seen in powder nanomaterials but less so in solids. However, nanomaterials are used to minimize these effects through controlled absorption [21]. Polymers possess unique qualities, including biocompatibility and biomimetic properties, which is why they are widely applicable in biomaterials as biosensors, catalysis, and the delivery of medicines [22]. Their use in medicines is beneficial due to their ability to distribute the therapeutic compound efficiently to the active site or target, making drug safety and efficacy certain [23].

2. Types of Polymer Based Nanocarriers

The precise classification of the nanocarriers is a complicated task to attempt. In the arena of biomedical and pharmaceutical nanotechnology, no specific boundaries have been defined, and therefore different perceptions may be relevant. However, a concise sketch of potential polymer-based nanocarriers is important as depicted in Figure 1. Depending on their origin they can also be identified as natural and synthetic polymeric nanoparticles. These are distinguished by their concurrent polymeric design and colloidal (1–1000 nm) measurements and have been commonly identified as polymeric (biodegradable and biocompatible) nanoparticles, dendrimers, nanoemulsions, polymersomes, polymeric micelles, or biopolymer complexes or cubosomes [24].
The highly relevant articles were retrieved via various search engines on the databases, Science Direct, Web of Sciences, PubMed, Scopus, PubChem, and Google Scholar. The examples have been selected on the basis of their significance to provide experimental insight into each type of nanocarriers to the readers. The keywords and phrases used for the search are “dendrimers”, “polymeric micelles”, “polymerosomes”, “nanogels”, “dendrimers”, “cubesomes”, “NLCs”, “nanographene”, “nanocomposites”, “CNTs”, “polymers”, and “nanocarriers”, “In vitro and in vivo activities”. The inclusion criteria implied for selecting particular polymer-based nanocarriers represent the significance of that particular study.

2.1. Polymer-Based Nanoparticles

Nanoparticles are sub-nanosized colloidal structures composed of synthetic or semisynthetic polymers. NPs range in size between 1 and 100 nm [25]. The ultra-small size of NPs exhibits a larger surface area to volume ratio and hence allows greater drug loading within the core via encapsulation and onto the surface via absorption. Their unique size range allows for extended circulation, leading to prolonged drug release. The entrapment of drugs in NPs protects against chemical degradation and improves their pharmacokinetic properties, leading to controlled release over a long period. NPs exhibits flexibility of surface functionalization with targeting ligands for site-specific drug delivery and thereby improve therapeutic efficacy [26]. Materials used in the manufacturing of nanocarriers based on polymers are selected not only for their functional characteristics, such as their capability to structure planned nanostructures, but also for their biological behavior. Polymers that are biodegradable and biocompatible are usually favored. Given the multitude of accessible or easily synthesizable polymers with promising potential for medicinal use, regulatory approval has been achieved for just a few. Various categories and forms of polymers including poly-lactic-co-glycolic acid (PLGA), poly-DL-lactic acid (PDLLA), polycaprolactone (PCL), polyacrylate, polymethacrylate, cellulose by-products, poly(ethylene oxide) triblock (PEO)/polypropylene oxide (PPO) (PEO-PPO-PEO; poloxamers), PEG, polyvinyl alcohol (PVA) and alginate was licensed by the United States Food and Drug Administration (USFDA) and other foreign authorities for various medical purposes over the years [27,28].
However, not all of these have sufficient features to develop nanomaterials that can act as nanocarriers, so unsurprisingly previously authorized materials may be standard candidates for rapid clearance by regulatory agencies. In comparison, modern polymer conjugates and polymers need rigorous biological and toxicological assessment to be approved for human use, and their clearance may be overwhelming [29].
Yaşar et al., 2020 developed polymer-based NPs with 74 nm particle size for selective protein recognition by using thiol-ene miniemulsion photopolymerization. The developed NPs revealed more selectivity toward myoglobin [30].
Puri et al., 2017 fabricated pH-sensitive polymer-based NPs to deliver bioactive compounds employing a dispersion polymerization approach. They reported that the rate of hydrolysis and drug discharge were quicker at pH 5.0 compared to pH 7.4 [31].

2.2. Polymeric Micelles

Fabricated by amphiphilic block copolymers, the polymeric micelles can be self-assembled (and disassembled) under a valid concentration or temperature environment as versatile spherical structures (i.e., nanosized core-shell molecules produced by self-association of amphiphilic block copolymers when exposed to an aqueous solvent). In nature, the core of micelles is hydrophobic and appropriate for incorporating hydrophobic moieties [32]. They are suitable for parenteral delivery compared to current solubilizing agents (e.g., cremophor EL), causing the dosage of effective but toxic and poorly water-soluble compounds to increase [33]. Investigations into nano-bio dynamics which can function at the molecular, cellular, and tissue levels is often ignored [34].
Varshosaz et al., synthesized docetaxel-loaded polymeric micelles using folic acid grafted synperonic PE/F 127-cholesteryl hemisuccinate copolymer for precise delivery to cancerous cells. The in vitro cellular uptake observations in B16F10 melanoma cells revealed improved uptake of targeted polymeric micelles through folic acid receptor-mediated endocytosis [35]. The micelles were found to be non-toxic and safe for biomedical usage over a range of 0.001 to 500 µg/mL. The morphological examination revealed the smooth spherical shape of the micelles. In vitro drug release investigation showed an initial burst trailed by slower and sustained release up to 134 h. The early burst release could be credited to the quick dissolution of surface-bound drugs followed by steady diffusion from the core of micelles. An in vivo antitumor efficiency study in B16F10 melanoma-bearing mice revealed effectively inhibited tumor growth by the drug-loaded targeted polymeric micelles compared to non-targeted polymeric micelles as shown in Figure 2.
Chida et al., fabricated epirubicin (EPI) loaded polymeric micelles armed with pH-sensitive drug release characteristics in contradiction to axillary lymph node metastasis (ALNM) of triple-negative breast cancer (TNBC) for improving the therapeutic effect. By accumulating selectively and penetrating both the vascularized ALNM and primary tumor, as well as effective drug trigger caused by intratumoral acidic atmosphere, the proposed formulation successfully prevented the growth of ALNM and the expansion of the primary tumor [36].
Bae et al., designed environment-sensitive supramolecular structures for intracellular drug delivery of Adriamycin (ADR) via linking it to the polymer by a pH-sensitive coupler. The micelles treated carcinoma cells via their transportation into a cell by endocytosis, consequently avoiding cell-membrane transporters [37].

2.3. Polymersomes

Polymersomes are considered the next era of nanomedicine for clinical diagnosis and therapies due to their potential for controlling structure and features. These are the most favorable tools for a biomedical utility such as diagnosis and controlled drug delivery due to the compartmentalization of polymersomes. These are engineered amphiphilic block copolymers or vesicles, but in this circumstance they contain structures made up of one or more bilayers enclosing an aqueous center. In all conditions, the active load of molecules can be dissolved, dispersed, or chemically bound to nanocarriers. Their size is an important parameter for intracellular uptake. They offer better mechanical features and provide better structural and colloidal stability than liposomes in aqueous media [38]. Their flow properties, cellular uptake, and immune regulation depend upon their shape since their shape affects interaction with cells. As a result, being able to precisely rearrange a polymersome and comprehending the mechanisms underlying the various forms of interconversions is critical [39]. These polymers are amphiphilic in aqueous solvents and organized so that their hydrophilic head groups point toward the aqueous phase while their lipophilic tails are confined within the membrane’s core. The phase characteristic of a carrier in a solvent determines whether it can be built into a membrane [40].
Polymermerosomes are presently being studied for the delivery of different probes for imaging target tissues/organs and cytotoxic medications to canceous cells and also for gene therapy [41]. Zavvar and co-workers developed gadolinium-based quantum dots (QDs) of targeted PEG-PCL nanopolymersomes as a diagnostic agent and doxorubicin (DOX) as an anti-cancer agent. The controlled release of encapsulated DOX was revealed in the drug release study and the formulation was found to be stable in physiologic conditions. Enhancement in both toxicity and cellular uptake (p < 0.05) was revealed in MTT and flow cytometry results. Overall, the study demonstrated the theranostic application of targeted polymersomes with minimization of side effects [42]. Zhou et al., developed polymersomes based on photo-response for delivery of doxorubicin hydrochloride. The drug can be entrapped into their hydrophilic voids and their photo-responsive disintegration resulted in a controlled release of the drug. The cellular assay demonstrated their active targeting of folic acid and photo-activated release with higher cytotoxicity of HeLa cells. The study revealed the potential of polymersomes as effective and intelligent drug carriers [43].
Li et al., designed paclitaxel-loaded vesicles from matrix metalloproteinase (MMP)-cleavable peptide-linked triblock copolymer, polyethylene glycol-GPLGVRG-b-poly(ε-caprolactone)-b-poly(3-guanidinopropyl methacrylamide) that showed considerably improved cellular internalization effectiveness (~10 folds) in contrast to its pure form. The developed vesicles exhibited greater cytotoxicity against MMP-overexpressing HT1080 cells and multicellular spheroids. When MMP-2 was used for treatment it resulted in cleavage of stealthy PEG shell. The vesicles undergo morphological transformation into fused multicavity vesicles and small nanoparticles, accompanied by a redistribution of PGPMA segments with 76% exposed to the outside [44].
To increase chemical reaction efficacy, Li et al., created polymeric nanoreactors (NRs). They created therapeutic vesicular NRs (theraNR) that were loaded with glucose oxidase (GOD). When injected into normal tissues, the theraNR was inactive. The tumor acidity activates them precisely at the target location. They effectively destroy cancer cells by inhibiting their antioxidant capacity and perfusing tumors through a synergistic action [45].
Li et al., created a polymersome nanoreactor based on a block copolymer prodrug that may enable new cancer oxidation/chemotherapy via selective activation at tumor locations. GOD-loaded polymersome nanoreactors were created by optimizing block copolymers to self-accumulate into polymersomes in an aqueous solution for encapsulation of GOD. Through high tumor oxidative stress, oxidation/chemotherapy and released CPT medicines synergistically killed cancerous cells and inhibited tumor development [46].
By simply combining a couple of differently charged block copolymers (poly(amino acid)s and PEG) in an aqueous medium, Koide et al., produced a PICsome (a novel form of polymer vesicle along with a polyion complex (PIC) membrane) with a size range of up to 10 µm. Semipermeable characteristic of PICsomes membrane was discovered using spectral analysis and confocal laser scanning microscopic imaging. Even in the existence of serum proteins, the PICsomes maintained a high level of physiological stability, indicating that they may be used in medical applications such as therapeutic vehicles [47].
By incorporating stimuli-sensitive linkers into a crosslinking membrane network, Li et al., created responsive nano-reactors built on polyion complex vesicles. GOD could be protected by the fabricated ROS-responsive nano-reactor with self-improving catalytic glucose oxidation, which is attributed to stimuli-reactive vesicle development short of breakage and size-selective payload release pattern, to attain cytocidal role by oxidative stress induction and glucose starvation. Pyroptosis, an immunostimulatory type of cell demise, was produced by the GOD-loaded therapeutic nanoreactor [48].

2.4. Dendrimers

Dendrimers (accurately designed, highly-radial polymeric frameworks) were incorporated as promising drug nanocarriers, although some authors seem to be more satisfactory in their description as “polymer therapeutics” [49]. These are polymeric nanomaterials having 3-D macromolecule having branch-like shape, with less than 15 nm of size and coming out from a central portion [50,51].
Pillay et al., developed folic acid loaded dendrimer-functionalized selenium nanoparticles (FA-PAMAM-SeNPs) with size <150 nm and zeta potential of >25 mV. The developed formulation demonstrated greater cell viability (>85%) in contrast to placebo nanocomplexes (75%), confirming the importance of selenium in the developed formulation. The developed formulation resulted in greater overall transgene expression that showed their improved receptor-mediated cellular uptake [52].
Bartusik-Aebisher et al., developed a trastuzumab-dendrimer-fluorine DDS in which API was attached to dendrimers (fluorinated) via covalent bonding. They found that the potential of the developed formulation was more efficient than pure trastuzumab [53].
Mota et al., encapsulated I-buthionine sulfoximine in folate-targeted polyurea dendrimers. Initially, the burst release was observed that was expected in these delivery systems. After 1 h, about 60% of drug release was found, reaching a plateau after 3 h. After 24 h, 90% drug release was found [54].
Zhang et al., 2022 fabricated a third generation of poly(phosphorhydratzone) radical dendrimers and reported their action against the diagnosis of brain tumors [55]. It was also reported that the developed agent did not cause any toxicity to the cells.
Knauer et al., 2022 designed a polycationic phosphorous dendrimer-based approach for the delivery of siRNA to target Lyn [56].

2.5. Nanosized Hydrogels

Recently, the advancement of biological science managed novel developments in the area of in vitro cell culturing. Various novel innovations such as cells on chips/cell microarray need consistent supportive materials with decent biocompatibility, and cell adhesion and are simple to use [57]. Nanomaterial-based systemic drug vehicles need various properties particularly delivery of drugs to targeted sites. These parameters increase ligand conjugation and passive targeting, resulting in an improvement in active targeting. Hydrogel-medicated local DDSs directly deliver drug moieties at the target site without complications due to their implantation proximate to carcinoma tissues. It has been already found that a visible light-cured injectable glycol chitosan (GC) hydrogel-based local DDS can be employed for an ailment of various solid cancers [58]. Nanosized hydrogels developed by physically or chemically cross-linking polymer systems and commonly called nanogels can also be known as nanocarriers based on polymers [59]. Hydrogels may be custom-shaped and thickened, and their surfaces can be shaped using lithography methods. Furthermore, hydrogels can be fabricated with biomolecules to achieve unique characteristics [58].
Despite their solute permeability and controllable release features, hydrogels are efficient carriers for medicines, proteins, and others. Hydrogels made of chitosan can create multilayered structures, which is why they demonstrate utility in distributing bioactive molecules including insulin and growth factors, and in cell and tissue organization. They can be administered to the body using very simple, minimal stress procedures for the administration such as injection and both ocular and nasal administration [60].

2.6. Polymeric Cubosomes

Polymeric cubosomes are newly discovered inverse bicontinuous cubic mesophases. These are colloidal particles containing mesoporous block copolymers having well-defined reticulated pore networks. Kim et al., prepared polymer cubosomes using block copolymer which was synthesized by linking hydrophilic PEG and hydrophobic polyisoprene polymer blocks. The cubosomes were prepared via solution self-assembly of block copolymers [61]. The block copolymers with non-linear structures undergo self-assembly to form inverse mesophases in solution [62]. These are highly efficient nanoparticles that are developed from a lipidic cubic state and protected by an outer layer of polymer. The polymeric cubosomes are considered to have improved endurance and have a greater capacity to surround and embody hydrophobic anticancer agents due to their liquid crystalline membrane structure [63,64,65,66,67]. As a result of recent developments, applications involving drug delivery, membrane bioreactor, artificial cells and biosensors can be developed in vitro in both bulk and nanoparticle formulations [68].
Zhang et al., developed the cubosomes loaded with a combination of anti-cancer drug cisplatin and paclitaxel for an effective targeting against cancer cells. In this work, a slow and sustained release was obtained by coating the cubosomes with poly-Ɛ-lysine. The coating prevented the initial burst of API resulting in a sustained drug release. TEM studies revealed very easily distinguishable spherical polyangular structures. Both initial reductions in burst discharge of drug and a slow, sustained discharge with time were revealed by in vitro studies. The cytotoxicity studies revealed the non-toxic behavior of cubosomes [69]. The morphological pattern, in vitro drug release, and cell viability investigations are shown in Figure 3. The impedance measurement and fluorescent imaging studies further confirmed their therapeutic efficiency against HeLa cells. Overall, the study affirms their significance as an effective nano-drug carrier.
Boge and co-workers developed the cubosomes for application in the delivery of peptide LL-37 for the ailment of skin infections. In the first strategy of development, the LL-37 was first added into a gel followed by dispersion into nanoparticles. The next strategy involved the adsorbing LL-37 onto preformed cubosomes. The last strategy consisted of the incorporation of LL-37 during their spontaneous formation in an ethanol/glycerol monooleate mixture. The pre-loaded cubosomes showed maximum efficiency in killing the bacteria [70].

3. Routes of Administration and Applications of Polymer Based Nanocarriers

The merging of polymer technology with pharmaceutical research resulted in a quantum leap in terms of innovation in designing and developing novel DDSs such as nanotechnology-based delivery systems. The application of nanopolymers in pharmaceutical and biomedical fields are swiftly growing, such as developing scaffold in tissue engineering, designing a drug delivery system, ophthalmology, dentistry, bone repair, implantation of medical devices, prosthesis, and many other medical fields. It is possible to obtain desirable properties in a pharmaceutical polymer by modifying its physical and chemical characteristics such as molecular weight, type, composition, co-polymerization, biomimetic, and co-processed excipient [71].
The discovery of the first synthetic drug delivery system sparked an interest in designing novel biodegradable polymers to replace the non-degradable polymer. The advent of bioadhesive polymer resulted in improved residence time and intimate contact between polymer and epithelial surface. Further advancements in polymer science led to the discovery of smart polymers that release the drug in presence of specific stimuli (physical, chemical, or biological) [72]. These nanopolymers were widely scrutinized based on their mechanism for controlled release of drugs and the understanding of their behavior in solid and liquid dosage forms, disperse systems, transdermal patches, implants, and many others [73]. There are numerous biodegradable and non-biodegradable polymers possessing different physicochemical characteristics used in pharmaceutical and biomedical fields (as shown in Table 1) [74,75,76,77,78,79,80,81,82,83,84].
Drug molecules (such as proteins, peptides, and DNA) are protected and stabilized by these polymers from environmental hazard degradation. Even though polymers have wide applications in the pharmaceutical industry, the criteria for proper selection and design of polymers is a challenging task and requires an in-depth understanding of its bulk properties (such as molecular weight, diffusion, and dissolution-controlled release based on solubility) and surface properties (such as surface energy, smoothness, hydrophilicity, lubricity, water sorption, and swellability) in addition to its physicochemical properties. The structure-activity relationship and polymeric matrices can be altered by changes in chemical composition and microstructural design [71]. The polymer-based nanocarriers are widely employed in various arenas for differenet routes that are discussed in brief below.

3.1. Applications of Polymeric Nanocarriers in Oral Drug Delivery

The oral route has been explored as a non-invasive and preferred delivery route for numerous therapeutic agents. However, oral delivery suffers poor bioavailability and enzymatic instability [85,86,87]. Modulation of gastrointestinal transit time is a challenge that can be overcome by increasing the residence time of drugs within the gastrointestinal tract. Polymeric nanocarriers emerged as prominent oral delivery vehicles owing to their interesting bioavailability, superior drug entrapment, control release, and minimal toxicity. The improved oral bioavailability of polymeric nanocarriers is due to the enhanced mucoadhesion property of polymer that sticks to the mucosa of the gastrointestinal tract leading to prolonged gastro-intestinal transit time and thereby enhancing mucosal penetration (as shown in Figure 4).
Polymeric materials being biocompatible, biodegradable, non-immunogenic, and functional that offer numerous advantages for drug delivery such as active drug targeting via surface functionalization and controlled release in the body [88]. Polymers-based nanocarriers have been extensively explored as vehicles for oral delivery of various peptides, genes, medicaments, proteins, siRNA, probiotics and vaccines as enlisted in Table 2 [89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110]. Polylactic acid (PLA), poly-glycolic acid (PGA), polycyanoacrylates (PCA), polyethyleneimine, and polycaprolactone are the main synthetic polymers widely employed for oral drug delivery. Chitosan, dextran, gelatin, alginate, and agar among which chitosan is most widely used owing to its mucoadhesive, biocompatible, and non-toxic properties [100].

3.2. Applications of Polymeric Nanocarriers in Ocular Drug Delivery

Several efforts have been made in ocular drug delivery to increase corneal assimilation by improving the drug residence time with the ocular surface through bioadhesive polymers [111]. The precorneal fluid contains a mucin-glycocalyx domain which is bound to the corneal surface and serves as a substrate for adequate binding of polymeric chains present in ocular formulation through non-covalent bonding resulting in prolonged drug contact time with the ocular surface and thereby improved bioavailability (as demonstrated in Figure 5). The widely used polymers for ocular drug delivery include chitosan, gelatin, alginate, synthetic poly-alkyl cyanoacrylate, poly-ε-caprolactone, poly-lactic acid, poly-lactic-co-glycolic acid, and polystyrene [112]. The biological properties of the polymer such as biodegradability, non-toxicity, biocompatibility, and mucoadhesiveness resulted in the development of various colloidal vehicles for ocular routes such as polymeric nanoparticles and polymeric micelles [113]. The positive surface charge of polymeric nanocarriers allows prolonged contact time of API on the ocular surface due to increased interaction with glycoprotein of cornea and conjunctiva leading to the formation of a precorneal depot that is responsible for the prolonged release of API.
Sharma et al., formulated polymeric nanoparticles using bioadhesive positively charged Eudragit® RS100 and Eudragit® RL100 which are official by USFDA as excipients for controlled drug delivery. The presence of a positive charge on nanoparticles resulted in ionic interaction with anionic mucin existing in the mucus layer of tear film leading to sustained release of drug and improved ocular penetration [114]. Mahor et al. fabricated moxifloxacin-loaded polymeric nanoparticles using positively charged gelatin for its effective ocular delivery and controlled release in the corneal eye layer [115]. Mittal et al. utilized chitosan, a polycationic bioadhesive polymer, to develop timolol maleate-loaded polymeric nanoparticles. The polymeric nanoparticles exhibited significant bioadhesive strength due to the existence of polymeric chains and polar functional groups in chitosan. The in vitro experiment displayed burst release (27.18% in 1 h) and then sustained release (90.55%) up to 24 h. The ex vivo transcorneal permeation investigation exhibited appreciable corneal penetration of timolol from polymeric nanoparticles compared to conventional eye drops attributable to the penetration-enhancing property of chitosan [116]. Table 3 compiles significant reports on the ocular delivery of polymeric nanocarriers [117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148].

3.3. Applications of Polymeric Nanocarriers in Parenteral Drug Delivery

This is the preferred route for delivery of active ingredients having a narrow therapeutic index and poor bioavailability. However, intramuscular and subcutaneous injection dosage form suffers rapid drug elimination. Continuous IV infusion is one of the approaches for maintaining constant drug delivery which also avoids hepatic metabolism but the patient has to be hospitalized. Among methodologies to overcome traditional medical treatment, simulation of IV infusion by use of parenteral controlled release formulations has gained interest. Figure 6 depicts sustained and targeted delivery of drugs via polymeric nanocarriers following parenteral administration [148].
Active targeting can be achieved at different levels depending on the extent of penetration; it may occur at the organ, cell, or subcellular level. It is important to note that in solid tumors, even active targeting processes begin with the passive accumulation of the DDS in the tumor tissue, so any actively targeted carrier must satisfy the basic requirements outlined for passively targeted systems: the polymer/nanoparticle should be biocompatible, and the system must be stable in circulation, long-circulating, and of a size that permits efficient extravasation and accumulation in tumors. In addition, actively targeted systems are often more effective in general than passively targeted alternatives [149].
Intravenous administration of biodegradable polymeric nanocarriers has been effectively employed for controlling and targeting drugs to a specific site of action. In particular, biodegradable nanocarriers formulated from PLGA have been extensively studied for sustained and targeted delivery of numerous agents [150]. It is a copolymer synthesized by co-polymerization of two different monomeric units, the cyclic dimers of glycolic acid and lactic acid, linked through an ester linkage. PLGA has been approved by FDA and European Medicine Agency for usage in parenteral DDSs due to its biocompatibility and biodegradability. Several forms of controlled DDSs have been studied among which polymeric nanocarriers have gained attention for diagnostics, prognostics, controlled and sustained delivery of protein, peptide, pDNA, and other therapeutic agents owing to their biocompatibility, non-immunogenicity, and several others are enlisted in Table 4 [151,152,153,154,155,156,157,158,159,160,161]. Natural and synthetic polymers have been extensively investigated as carriers for several anticancer drugs. Conjugation of drugs with these polymeric carriers increases their circulation time in the blood leading to passive gathering in tumor tissues due to the enhanced permeability and retention (EPR) effect [140,141].

3.4. Applications of Polymeric Nanocarriers in Topical Drug Delivery

Polymer-based nanocarriers have been extensively explored as topical formulations to improve cutaneous delivery owing to their physicochemical attributes which prevent degradation or denaturation of unstable drugs, reduce their toxic side effects through controlled release, and enhance their cutaneous penetration across the biological membrane more precisely skin barrier due to their ability to increase the concentration gradient [162]. Polymers serve an important role in the preparation of polymeric nanocarriers which might be natural or synthetic. The extensively used natural polymers are chitosan, alginate, gelatin, and albumin. Synthetic polymers include polylactides, PVA, poly(acrylic acid), polyacrylamide, PEG, and several others. These polymers have gained attention as a substrate for colloidal vehicles due to their blood stability, non-toxicity, non-thrombogenic, non-immunogenic, and non-inflammatory characteristics [163,164].
Natural polymers tend to develop hydrogels which render them suitable carriers for peptides, oligonucleotides, proteins, and water-soluble APIs for improving the absorption and targeted drug delivery of polymeric nanocarriers for topical administration. Chitosan-based nanocarriers have been widely used among the natural polymers for topical delivery. An N-deacetylated derivative of chitin, chitosan is a cationic polymer that is made up of glucosamine units. Its additional anti-oxidant, anti-inflammatory, and anti-microbial features make it an appropriate carrier for therapeutic delivery. Table 5 reports a compilation of polymeric nanocarriers for topical delivery of drugs, peptides, and genes [148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165].

4. Practical and Regulatory Consideration

Nanomedicine is a new area integrating nanotechnology with pharmaceutical and biomedical sciences, intending for the development of medicines with improved safety and toxicological profiles [171]. In drug formulation, nanoparticles can impart several physical and biological advantages such as improved pharmacokinetic profile, enhanced tissue/organ selectivity, and decreased toxicity as compared to conventional medicines [172]. FDA governs the pre-market approval process of conventional drugs, biologics, and medical devices, and likewise, nanomedicines are also subject to a normal range of pre-clinical and clinical validation. Based on available literature and the Centre for Drug Evaluation and Research (CDER), the no. of clinical trials involving nanomedicines has increased three-fold and the utility of nanomaterials in these products has incredibly risen in the past two decades [173]. Based on previously reported results, liposomes account for the largest proportion of therapeutic nanoproducts with over 33%, followed by nanocrystals and emulsions which accounts for 23% and 14%, respectively. Nine percent of products used include polymer ion complexes while micelles have a 6% share. The studies also show that specific nanostructures (quantum dots, carbon nanotubes, graphene) also have a small percentage of shares in these products [174].
In nanomedicine, polymeric nanoparticles serve two purposes (i.e., controlled release of drug-using biodegradable polymers and improved half-life and bioavailability by forming drug-polymer conjugates) [175]. Copaxone and Neulasta are two polymeric drugs out of the top 10 bestselling drugs in the US in 2013 [176]. There are several polymeric immobilized nanomedicines as shown in Table 6 that are approved by USFDA [177,178,179,180,181,182,183,184].
The composition and structure of polymeric micelles can be precisely modified to achieve specific release characteristics and drug loading. The only approved polymeric micelle is a traditional formulation of oestradiol (Estrasorb™). The use of modern co-block polymers led to the development of polymeric micelles at a lower critical micellar concentration with high stability [185]. Few polymeric micelles formulations that are in late-stage clinical trials, especially i/v administered formulations are shown in Table 7 [186].
The various patented reported polymeric nanocarriers intended for oral, ocular, topical, and parenteral administration are enlisted in Table 8 [187].

4.1. Toxicity Issues

Most of the polymeric nanocarriers are generally regarded as safe owing to the interesting physicochemical characteristics of the polymer which render them biocompatible and biodegradable. Copolymers PLA and PLGA acid are the most extensively employed biomaterials for polymeric nanocarriers, that undergo hydrolytic degradation within the body leading to the generation of endogenous lactic acid and glycolic acid monomers which are metabolized via Kreb’s cycle leading to minimal systemic toxicity [188]. However, various factors could affect the toxicity of the polymeric nanocarriers not limited to their surface chemistry, size, and concentration as reported by Ahlawat and Henriquez [189]. The size of the polymeric nanocarriers significantly affects the cellular uptake and hence causes toxicity. Enhanced cellular uptake of polymeric nanocarriers leads to increased interaction with intracellular components which triggers the production of free radicals, cytokines, and reactive oxygen species (ROS) causing organelle destruction and finally cell lysis that leads to toxicity issues [190,191].
Grabowski and Hillaireau studied the toxicity of surface-mediated polymeric nanoparticles on human-like THP-1 macrophages wherein cell mitochondrial activity, apoptosis detection, ROS, and pro-inflammatory cytokines production were chosen as toxicity criteria. Positively, negatively and neutral PLGA nanoparticles were fabricated with chitosan, poloxamer 188, and PVA stabilizers. The stabilizer-free PLGA nanoparticles did not exhibit any sign of toxicity at or above a therapeutic concentration of 0.1 mg/mL. However, PLGA nanoparticles prepared using stabilizers exhibited cytotoxicity above the therapeutic level of 0.1 mg/mL, thereby confirming the safety of the polymeric nanocarriers and the toxicological contribution of stabilizers used in the polymeric nanocarrier formulation [192].
Rejinold and Muthunarayanan reported non-toxicity of the chitosan-g-poly-N-vinyl caprolactone nanoparticles in an array of cell lines at 100–1000 µg/mL [193]. Similarly, Dandekar and Dhumal reported the cellular safety of the Eudragit® S100 polymeric nanoparticles through genotoxicity studies [194]. Singh and Ramarao examined the concentration-dependent toxicity of PLGA, PLA, poly-ϵ-caprolactone, and poly-lactide-caprolactone nanoparticles using macrophage (RAW 264.6), hepatocytes (Hep G2), lung (A549), kidney (A498) and neuronal epithelial (Neuro A2) [180]. Polymeric nanoparticles did not display toxicity at 100 µg/mL concentration as marked by no change in cell viability. However, a reduction in cell viability was detected at 300–1000 µg/mL concentration after 72 h incubation of polymeric nanoparticles with cell lines.

4.2. Effects of the Route of Administration on Bioavailability

Polymeric nanocarriers have become a prominent field of research in the arena of drug delivery since they can improve pharmacokinetic and bioavailability that results in effectiveness of various drugs, enhanced hydrophilicity, reduction of interaction with plasma and cellular proteins, and greater desposition of drug at target sites [195]. Oral, intravenous, intramuscular, intranasal, intradermal, and transdermal administration are the foremost delivery routes for drugs. Other routes, such as ocular delivery, have also been developed for localized and targeted drug delivery, ocular route is employed that declines unwanted systemic side effects. Specific barriers are encountered by each route of administration route. Roughly, solubility and permeability are two major issues related to drug substances that are responsible for bioavailability upon administration [196]. Improved stability and bioavailability of therapeutic agents coupled with extended drug release profile and possibility to embed numerous drugs makes suitable candidate for pharmaceutical products [197].
Various factors govern the assimilation of the drug such as drug solubility, mucosal permeability, and stability in the GI tract milieu. Efforts are ongoing to tackle these issues have focused on physicochemical, biochemical, biological, and metabolic barriers that are responsible for limited assimilation. Different pharmaceutical technologies and DDSs such as nanocarriers, micelles, cyclodextrins, and lipid-based carriers have been discovered for improving the assimilation of a drug.
GI tract leads to experience a varied range of pH, enzymatic degradation and low permeability across the membrane. Polymeric NPs may be an auspicious strategy that can improve stability to crossing the mucus barrier or interact with a mucus layer and increase residence and contact time [198].
Oral, aerosol, and nasal vaccination is a novel immunization technique that employ nanocarriers to administer vaccine. Therapeutic agents and vaccines with lower penetration potential can be administer via transmucosal route while maintaining their biological activity. Nowdays, nanocarriers for transmucosal delivery are gaining more attention since delivery via oral mucosa helps evade first-pass metabolism, improves bioavailability and acts as a means of rapid drug transport to the systemic circulation. The oral transmucosal vaccine delivery by nanocarriers is the most upcoming innovation in efficient vaccine delivery [199,200].

5. Future Prospects

Over the past few decades, polymer science has evolved to mediate safe and efficient drug delivery for the ailment of several health conditions. Over the years, the usage of polymers has been limited to oral delivery, but critical advances in polymer science such as the development of biodegradable nanosized carriers have accelerated their use in parenteral formulations. Further innovations in the application of polymers have led to a more sophisticated approach, thereby improving the pharmacokinetics, decreased toxicity, stealth effect, cell or organ-specific targeting, and controlled release of the drugs; these innovatioms have made it possible to administer two or more drugs or imaging/diagnostic agents within the same drug delivery system, leading to more effective therapy [198,201,202]. Due to the development of novel polymers with highly complex structures and functions, its scope has widened in the field of nanoformulation, particularly in imaging and theranostic applications [203]. The recent trend in polymer science is leaping from the traditional linear polymer to the complex hyperbranched or hybrid polymers.
New technologies and advances in the synthesis and manufacturing process such as 3-D printing, supercritical fluid technology, and nanoengineering are revolutionizing polymer-based drug delivery systems [204]. Scale-up in the field of nanotechnology has always been troublesome due to non-reproducibility between batches and future evolution is expected from the introduction of new types of polymers also known as smart polymers (stimuli-responsive, enzyme responsive) that fulfill requirements to be used as a constituent in drug delivery systems [205]. Nevertheless, future development should progress towards understanding the interaction of polymers or polymercarriers with the biological system. Most studies carried out in the present scenario neglect the biocompatibility studies that fail new devices at the later stage of development. Adequate animal models and in vitro studies can lead to a breakthrough in polymer science as safe and effective platforms for drug delivery [206].
Nanomedicine has evolved over the last several decades from biologically inert substances to increasingly intelligent systems aiming at improving in vivo functioning. But, we must acknowledge that most systems depend on logical explanations-including some over-explanation rather than conclusive proof, which is a pivotal event in nanomedicine’s progress. The importance of investigating nano-bio interactions and desired functioning at the molecular, cellular, and tissue levels is often ignored [34].
An overall change in global markets, technological innovations, and data on the pathophysiological and cell-based mechanisms should be considered that can outline the future progress for polymers in drug delivery.

6. Conclusions

Polymeric nanocarriers have a great potential for clinical applications. They are expected to perform both diagnostic and therapeutic functions due to their interesting physiochemical characteristics which offer numerous advantages such as biocompatibility, biodegradation, non-immunogenicity, non-inflammatory, low toxicity, prolonged drug release, and active site targeting, rendering them safe for therapeutic applications at a certain concentration. At present, few polymeric systems can be employed for diagnostic and therapeutic applications. Among nanomaterials, polymeric nanocarriers have been the exceedingly explored delivery vehicles due to their wide range of solubility (hydrophilic and lipophilic surfaces) and self-assembling properties. They offer solubilization of hydrophobic or poorly soluble molecules, sustained drug release, and protection to encapsulated drugs from degradation and metabolism. The polymeric nanocarriers are considered as the best ones due to their good drug loading capacities, biodegradability, and stimuli-responsive control.
Polymeric nanocarriers are safe for the parenteral route of administration than their surfactant counterparts. They are kinetically stable and hence dissociate very slowly in blood, resulting in extended circulation time. Recently biopolymeric nanocarriers have immerged as storms which are developed using polymers obtained from natural sources. These biopolymeric nanocarriers offer better biodegradability, biocompatibility and could be the next generation nanocarriers for drug delivery. In the literature, there are several studies for developing new generation polymeric nanocarriers to obtain future smart and multifunctional nanomedicine.
Polymeric nanocarriers have many challenges for a carrier system such as biocompatibility, biodistribution, side effects, and biological barriers. Their biodegradability and biocompatibility profiles should be critically investigated before utilization in human clinical trials. One major concern that requires immediate attention is the regulatory concerns regarding the approval of polymer-based carriers since establishing new guidelines and policies seem to be in the initial stages of development. New criteria for manufacturing, testing, and storage of these nanosized polymeric materials should be defined by the regulatory agencies [207]. So, their manufacturing and regulation have challenges for their further development in various fields.

Author Contributions

A.K.T.—Conceptualization, Writing—Original Draft Preparation; S.C.U.—Writing—Review & Editing; M.K.—Conceptualization, Supervision; K.P.—Writing—Review & Editing, Data Curation; D.K.—Writing—Review & Editing, Data Curation; R.V.—Writing—Review & Editing; S.B.—Writing—Review & Editing; M.H.R.—Writing—Review & Editing. E.E.S.M. and S.C.—Writing—Review & Editing. 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

All the associated data are available within the manuscript.

Acknowledgments

The author would like to express his gratitude to the Deanship of Scientific Research at King Khalid University, Abha, Saudi Arabia, for funding through Research Groups Program under Grant No. R.G.P.2/7/43.

Conflicts of Interest

The authors declare that they have no conflict of interest.

Abbreviations

DOXDoxorubicin
NSMNanostructured materials
PCApolycyanoacrylates
PCLPolycaprolactone
PDLLAPoly-DL-lactic acid
PEGPolyethylene glycol
PGApoly-glycolic acid
PLAPolylactic acid
PLGAPoly-lactic-co-glycolic acid
PVAPolyvinyl alcohol
QDsQuantum dots
USFDAUnited States Food and Drug Administration.

References

  1. Available online: https://www.brainkart.com/article/Historical-development-of-polymers_6358/ (accessed on 3 July 2022).
  2. Batool, S.A.; Ahmad, K.; Irfan, M.; Ur Rehman, M.A. Zn-Mn-doped mesoporous bioactive glass nanoparticle-loaded zein coatings for bioactive and antibacterial orthopedic implants. J. Funct Biomater. 2022, 13, 97. [Google Scholar] [CrossRef] [PubMed]
  3. Hyldbakk, A.; Mørch, Y.; Snipstad, S. Identification of novel cyanoacrylate monomers for use in nanoparticle drug delivery systems prepared by miniemulsion polymerisation—A multistep screening approach. Int. J. Pharm. X 2022, 4, 100124. [Google Scholar] [CrossRef] [PubMed]
  4. Weissig, V.; Pettinger, T.K.; Murdock, N. Nanopharmaceuticals (part 1): Products on the market. Int. J. Nanomed. 2014, 9, 4357–4373. [Google Scholar] [CrossRef] [PubMed]
  5. Suk, J.S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L.M. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 2016, 99, 28–51. [Google Scholar] [CrossRef]
  6. Ahmed, M.; Lukyanov, A.N.; Torchilin, V.; Tournier, H.; Schneider, A.N.; Goldberg, S.N. Combined radiofrequency ablation and adjuvant liposomal chemotherapy: Effect of chemotherapeutic agent, nanoparticle size, and circulation time. J. Vasc. Interv. Radiol. 2005, 16, 1365–1371. [Google Scholar] [CrossRef] [PubMed]
  7. Gabizon, A.; Shmeeda, H.; Barenholz, Y. Pharmacokinetics of pegylated liposomal Doxorubicin: Review of animal and human studies. Clin. Pharmacokinet. 2003, 42, 419–436. [Google Scholar] [CrossRef] [PubMed]
  8. Laginha, K.M.; Verwoert, S.; Charrois, G.J.; Allen, T.M. Determination of doxorubicin levels in whole tumor and tumor nuclei in murine breast cancer tumors. Clin. Cancer Res. 2005, 11, 6944–6949. [Google Scholar] [CrossRef]
  9. Aghemo, A.; Rumi, M.G.; Colombo, M. Pegylated interferons α2a and α2b in the treatment of chronic hepatitis C. Nature Rev. Gastroenterol. Hepatol. 2010, 7, 485–494. [Google Scholar] [CrossRef]
  10. Miele, E.; Spinelli, G.P.; Miele, E.; Tomao, F.; Tomao, S. Albumin-bound formulation of paclitaxel (Abraxane® ABI-007) in the treatment of breast cancer. Int. J. Nanomed. 2009, 4, 99–105. [Google Scholar] [CrossRef]
  11. Werner, M.E.; Cummings, N.D.; Sethi, M.; Wang, E.C.; Sukumar, R.; Moore, D.T.; Wang, A.Z. Preclinical evaluation of Genexol-PM, a nanoparticle formulation of paclitaxel, as a novel radiosensitizer for the treatment of non-small cell lung cancer. Int. J. Radiat. Oncol. Biol. Phys. 2013, 86, 463–468. [Google Scholar] [CrossRef] [Green Version]
  12. Available online: https://clinicaltrials.gov/ct2/show/NCT00910741 (accessed on 18 January 2022).
  13. Hamaguchi, T.; Tsuji, A.; Yamaguchi, K.; Takeda, K.; Uetake, H.; Esaki, T.; Amagai, K.; Sakai, D.; Baba, H.; Kimura, M.; et al. A phase II study of NK012, a polymeric micelle formulation of SN-38, in unresectable, metastatic or recurrent colorectal cancer patients. Cancer Chemother. Pharmacol. 2018, 82, 1021–1029. [Google Scholar] [CrossRef] [PubMed]
  14. Available online: https://clinicaltrials.gov/ct2/show/NCT01644890 (accessed on 18 January 2022).
  15. Sinha, R.; Kim, G.J.; Nie, S.; Shin, D.M. Nanotechnology in cancer therapeutics: Bioconjugated nanoparticles for drug delivery. Mol. Cancer Ther. 2006, 5, 1909–1917. [Google Scholar] [CrossRef] [PubMed]
  16. Blasco, C.; Pico, Y. Determining nanomaterials in food. Tends Anal. Chem. 2011, 30, 84–99. [Google Scholar] [CrossRef]
  17. Nikam, A.N.; More, M.P.; Pandey, A.P.; Patil, P.O.; Patil, A.G.; Deshmukh, P.K. Design and development of thiolated graphene oxide nanosheets for brain tumor targeting. Int. J. Polym. Mater. 2020, 69, 611–621. [Google Scholar] [CrossRef]
  18. Verma, R.; Kaushik, A.; Almeer, R.; Rahman, M.H.; Abdel-Daim, M.M.; Kaushik, D. Improved pharmacodynamic potential of rosuvastatin by self-nanoemulsifying drug delivery system: An in vitro and in vivo evaluation. Int. J. Nanomed. 2021, 16, 905–924. [Google Scholar] [CrossRef]
  19. Verma, R.; Kaushik, D. Design and optimization of candesartan loaded self-nanoemulsifying drug delivery system for improving its dissolution rate and pharmacodynamic potential. Drug Deliv. 2020, 27, 756–771. [Google Scholar] [CrossRef]
  20. Patra, J.K.; Das, G.; Fraceto, L.F. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef] [PubMed]
  21. Gunasekaran, T.; Haile, T.; Nigusse, T.; Dhanaraju, M.D. Nanotechnology: An effective tool for enhancing bioavailability and bioactivity of phytomedicine. Asian Pacific J. Trop. Biomed. 2014, 4, S1–S7. [Google Scholar] [CrossRef]
  22. De, R.; Mahata, M.K.; Kim, K.T. Structure-based varieties of polymeric nanocarriers and influences of their physicochemical properties on drug delivery profiles. Adv. Sci. 2022, 9, e2105373. [Google Scholar] [CrossRef]
  23. Simon, V.; Cavalu, S.; Simon, S.; Mocuta, H.; Vanea, E.; Prinz, M.; Neumann, M. Surface functionalisation of sol-gel derived aluminosilicates in simulated body fluids. Solid State Ionics 2009, 180, 764–769. [Google Scholar] [CrossRef]
  24. Nicolas, J.; Mura, S.; Brambilla, D.; Mackiewicz, N.; Couvreur, P. Design, functionalization strategies and biomedical applications of targeted biodegradable/biocompatible polymer-based nanocarriers for drug delivery. Chem. Soc. Rev. 2013, 42, 1147–1235. [Google Scholar] [CrossRef] [PubMed]
  25. De Jong, W.H.; Borm, P.J. Drug delivery and nanoparticles:applications and hazards. Int. J. Nanomed. 2008, 3, 133–149. [Google Scholar] [CrossRef] [PubMed]
  26. Peracchia, M.T.; Fattal, E.; Desmaele, D. Stealth PEGylated polycyanoacrylate nanoparticles for intravenous administration and splenic targeting. J. Control. Release 1999, 60, 121–128. [Google Scholar] [CrossRef]
  27. Rowe, R.C.; Sheskey, P.J.; Quinn, M.E. Handbook of Pharmaceutical Excipients, 6th ed.; Pharmaceutical Press: London, UK, 2009. [Google Scholar]
  28. Inactive Ingredient Search for Approved Drug Products, U.S. Food & Drug Administration. 2020. Available online: https://www.accessdata.fda.gov/scripts/cder/iig/index.cfm, (accessed on 6 March 2020).
  29. Bao, Y.; Maeki, M.; Ishida, A.; Tani, H.; Tokeshi, M. Preparation of size-tunable sub-200 nm PLGA-based nanoparticles with a wide size range using a microfluidic platform. PLoS ONE 2022, 17, e0271050. [Google Scholar] [CrossRef] [PubMed]
  30. Yaşar, M.; Yöntem, F.D.; Kahraman, M.V.; Apohan, N.K.; Aktaş, Z.; Öncül, M.O.; Akçakaya, H. Polymeric nanoparticles for selective protein recognition by using thiol-ene miniemulsion photopolymerization. J. Biomater. Sci. Polym. Ed. 2020, 31, 2044–2059. [Google Scholar] [CrossRef]
  31. Puri, R.; Berhe, S.A.; Akala, E.O. pH-Sensitive polymeric nanoparticles fabricated by dispersion polymerization for the delivery of bioactive agents. Pharm. Nanotechnol. 2017, 5, 44–66. [Google Scholar] [CrossRef]
  32. Popovici, C.; Popa, M.; Sunel, V.; Atanase, L.I.; Ichim, D.L. Drug delivery systems based on pluronic micelles with antimicrobial activity. Polymers 2022, 14, 3007. [Google Scholar] [CrossRef]
  33. Perumal, S.; Atchudan, R.; Lee, W. A review of polymeric micelles and their applications. Polymers 2022, 14, 2510. [Google Scholar] [CrossRef]
  34. Li, J.; Kataoka, K. Chemo-physical strategies to advance the in vivo functionality of targeted nanomedicine: The next generation. J. Am. Chem. Soc. 2021, 143, 538–559. [Google Scholar] [CrossRef]
  35. Varshosaz, J.; Taymouri, S.; Hassanzadeh, F.; Javanmard, S.H.; Rostami, M. Folated synperonic-cholesteryl hemisuccinate polymeric micelles for the targeted delivery of docetaxel in melanoma. Biomed. Res. Int. 2015, 2015, 746093. [Google Scholar] [CrossRef] [Green Version]
  36. Chida, T.; Miura, Y.; Cabral, H.; Nomoto, T.; Kataoka, K.; Nishiyama, N. Epirubicin-loaded polymeric micelles effectively treat axillary lymph nodes metastasis of breast cancer through selective accumulation and pH-triggered drug release. J. Control. Release 2018, 292, 130–140. [Google Scholar] [CrossRef] [PubMed]
  37. Bae, Y.; Fukushima, S.; Harada, A.; Kataoka, K. Design of environment-sensitive supramolecular assemblies for intracellular drug delivery: Polymeric micelles that are responsive to intracellular pH change. Angew. Chem. Int. Ed. Engl. 2003, 42, 4640–4643. [Google Scholar] [CrossRef] [PubMed]
  38. Leong, J.; Teo, J.Y.; Aakalu, V.K.; Yang, Y.Y.; Kong, H. Engineering polymersomes for diagnostics and therapy. Adv. Healthc. Mater. 2018, 7, e1701276. [Google Scholar] [CrossRef]
  39. Men, Y.; Li, W.; Tu, Y.; Peng, F.; Janssen, G.A.; Nolte, R.J.M.; Wilson, D.A. Nonequilibrium reshaping of polymersomes via polymer addition. ACS Nano. 2019, 13, 12767–12773. [Google Scholar] [CrossRef]
  40. Miere, F.; Fritea, L.; Cavalu, S.; Vicas, S.I. Formulation, characterization and advantages of using liposomes in multiple therapies. Pharmacophore 2020, 11, 1–12. Available online: https://pharmacophorejournal.com/WFzrTpR (accessed on 18 January 2022).
  41. Gao, C.; Qi, S.; Liu, K. Functional characterization of Brassica napus DNA topoisomerase Iα-1 and its effect on flowering time when expressed in Arabidopsis thaliana. Biochem. Biophys. Res. Commun. 2017, 486, 124–129. [Google Scholar] [CrossRef]
  42. Zavvar, T.; Babaei, M.; Abnous, K. Synthesis of multimodal polymersomes for targeted drug delivery and MR/fluorescence imaging in metastatic breast cancer model. Int. J. Pharm. 2020, 578, 119091. [Google Scholar] [CrossRef]
  43. Zhou, Y.; Chen, R.; Yang, H.; Bao, C.; Fan, J.; Wang, C.; Lin, Q.; Zhu, L. Light-responsive polymersomes with a charge-switch for targeted drug delivery. J. Mater. Chem. B. 2020, 8, 727–735. [Google Scholar] [CrossRef]
  44. Li, J.; Xiao, S.; Xu, Y.; Zuo, S.; Zha, Z.; Ke, W.; He, C.; Ge, Z. Smart asymmetric vesicles with triggered availability of inner cell-penetrating shells for specific intracellular drug delivery. ACS Appl Mater. Interfaces 2017, 9, 17727–17735. [Google Scholar] [CrossRef]
  45. Li, J.; Dirisala, A.; Ge, Z.; Wang, Y.; Yin, W.; Ke, W.; Toh, K.; Xie, J.; Matsumoto, Y.; Anraku, Y.; et al. vesicular nanoreactors with tumor-specific activation and self-destruction for synergistic tumor ablation. Angew. Chem. Int. Ed. Engl. 2017, 56, 14025–14030. [Google Scholar] [CrossRef]
  46. Li, J.; Li, Y.; Wang, Y.; Ke, W.; Chen, W.; Wang, W.; Ge, Z. Polymer prodrug-based nanoreactors activated by tumor acidity for orchestrated oxidation/chemotherapy. Nano Lett. 2017, 17, 6983–6990. [Google Scholar] [CrossRef] [PubMed]
  47. Koide, A.; Kishimura, A.; Osada, K.; Jang, W.D.; Yamasaki, Y.; Kataoka, K. Semipermeable polymer vesicle (PICsome) self-assembled in aqueous medium from a pair of oppositely charged block copolymers: Physiologically stable micro-/nanocontainers of water-soluble macromolecules. J. Am. Chem. Soc. 2006, 128, 5988–5989. [Google Scholar] [CrossRef] [PubMed]
  48. Li, J.; Anraku, Y.; Kataoka, K. Self-boosting catalytic nanoreactors integrated with triggerable crosslinking membrane networks for initiation of immunogenic cell death by pyroptosis. Angew. Chem. Int. Ed. Engl. 2020, 59, 13526–13530. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, J.; Li, B.; Qiu, L.; Qiao, X.; Yang, H. Dendrimer-based drug delivery systems: History, challenges, and latest developments. J. Biol Eng. 2022, 16, 18. [Google Scholar] [CrossRef] [PubMed]
  50. Mittal, P.; Saharan, A.; Verma, R. Dendrimers: A new race of pharmaceutical nanocarriers. Biomed. Res. Int. 2021, 2021, 8844030. [Google Scholar] [CrossRef] [PubMed]
  51. Bober, Z.; Bartusik-Aebisher, D.; Aebisher, D. Application of dendrimers in anticancer diagnostics and therapy. Molecules 2022, 27, 3237. [Google Scholar] [CrossRef]
  52. Pillay, N.S.; Daniels, A.; Singh, M. Folate-targeted transgenic activity of dendrimer functionalized selenium nanoparticles in vitro. Int. J. Mol. Sci. 2020, 21, 7177. [Google Scholar] [CrossRef]
  53. Bartusik-Aebisher, D.; Chrzanowski, G.; Bober, Z.; Aebisher, D. An analytical study of Trastuzumab-dendrimer-fluorine drug delivery system in breast cancer therapy in vitro. Biomed. Pharmacother. 2021, 133, 111053. [Google Scholar] [CrossRef]
  54. Mota, P.; Pires, R.F.; Serpa, J.; Bonifácio, V.D.B. l-buthionine sulfoximine detection and quantification in polyurea dendrimer nanoformulations. Molecules 2019, 24, 3111. [Google Scholar] [CrossRef]
  55. Zhang, S.; Lloveras, V.; Lope-Piedrafita, S.; Calero-Pérez, P.; Wu, S.; Candiota, A.P.; Vidal-Gancedo, J. Metal-free radical dendrimers as MRI contrast agents for glioblastoma diagnosis: Ex vivo and in vivo approaches. Biomacromolecules 2022, 23, 2767–2777. [Google Scholar] [CrossRef]
  56. Knauer, N.; Arkhipova, V.; Li, G.; Hewera, M.; Pashkina, E.; Nguyen, P.H.; Meschaninova, M.; Kozlov, V.; Zhang, W.; Croner, R.S.; et al. In vitro validation of the therapeutic potential of dendrimer-based nanoformulations against tumor stem cells. Int. J. Mol. Sci. 2022, 23, 5691. [Google Scholar] [CrossRef] [PubMed]
  57. Chabria, Y.; Duffy, G.P.; Lowery, A.J.; Dwyer, R.M. Hydrogels: 3D drug delivery systems for nanoparticles and extracellular vesicles. Biomedicines 2021, 9, 1694. [Google Scholar] [CrossRef] [PubMed]
  58. Yoon, S.J.; Moon, Y.J.; Chun, H.J.; Yang, D.H. Doxorubicin·hydrochloride/cisplatin-loaded hydrogel/nanosized (2-hydroxypropyl)-beta-cyclodextrin local drug-delivery system for osteosarcoma treatment in vivo. Nanomaterials 2019, 9, 1652. [Google Scholar] [CrossRef] [PubMed]
  59. Szwajca, A.; Juszczyńska, S.; Jarzębski, M.; Baryła-Pankiewicz, E. Incorporation of fluorescent fluorinated methacrylate nano-sized particles into chitosan matrix formed as a membranes or beads. Polymers 2022, 14, 2750. [Google Scholar] [CrossRef]
  60. Liu, L.; Gao, Q.; Lu, X.; Zhou, H. In situ forming hydrogels based on chitosan for drug delivery and tissue regeneration. Asian J. Pharm. Sci. 2016, 11, 673–683. [Google Scholar] [CrossRef]
  61. Kim, J.; Yoon, M.; Jin, S.M.; Lee, J.; La, Y.; Lee, E.; Kim, K.T. Polymer cubosomes of block copolymers having cross-linkable soft hydrophobic blocks. Polym. Chem. 2019, 10, 3778–3785. [Google Scholar] [CrossRef]
  62. Sungmin, H.A.; Yunju, L.A.; Kyoung, T.K. Polymer cubosomes: Infinite cubic mazes and possibilities. Acc. Chem. Res. 2020, 53, 620–631. [Google Scholar] [CrossRef]
  63. Boge, L.; Bysell, H.; Ringstad, L. Lipid-based liquid crystals as carriers for antimicrobial peptides: Phase behavior and antimicrobial effect. Langmuir 2016, 32, 4217–4228. [Google Scholar] [CrossRef]
  64. Angelova, A.; Garamus, V.M.; Angelov, B.; Tian, Z.; Li, Y.; Zou, A. Advances in structural design of lipid-based nanoparticle carriers for delivery of macromolecular drugs, phytochemicals and anti-tumor agents. Adv. Colloid Interface Sci. 2017, 249, 331–345. [Google Scholar] [CrossRef]
  65. Zerkoune, L.; Lesieur, S.; Putaux, J.L. Mesoporous self-assembled nanoparticles of biotransesterified cyclodextrins and nonlamellar lipids as carriers of water-insoluble substances. Soft Matter. 2016, 12, 7539–7550. [Google Scholar] [CrossRef]
  66. Zou, A.; Li, Y.; Chen, Y. Self-assembled stable sponge-type nanocarries for Brucea javanica oil delivery. Colloids Surf. B Biointerfaces 2017, 153, 310–319. [Google Scholar] [CrossRef] [PubMed]
  67. Wakaskar, R.R. General overview of lipid-polymer hybrid nanoparticles, dendrimers, micelles, liposomes, spongosomes and cubosomes. J. Drug Target. 2018, 26, 311–318. [Google Scholar] [CrossRef] [PubMed]
  68. Nasr, M.; Saber, S.; Bazeed, A.Y.; Ramadan, H.A.; Ebada, A.; Ciorba, A.L.; Cavalu, S.; Elagamy, H.I. Advantages of Cubosomal Formulation for Gatifloxacin Delivery in the Treatment of Bacterial Keratitis: In Vitro and In Vivo Approach Using Clinical Isolate of Methicillin-Resistant Staphylococcus aureus. Materials 2022, 15, 3374. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, L.; Li, J.; Tian, D.; Sun, L.; Wang, X.; Tian, M. Theranostic combinatorial drug-loaded coated cubosomes for enhanced targeting and efficacy against cancer cells. Cell Death Dis. 2020, 11, 1. [Google Scholar] [CrossRef]
  70. Boge, L.; Hallstensson, K.; Ringstad, L.; Johansson, J.; Andersson, T.; Davoudi, M.; Larsson, P.T.; Mahlapuu, M.; Håkansson, J.; Andersson, M. Cubosomes for topical delivery of the antimicrobial peptide LL-37. Eur. J. Pharm. Biopharm. 2019, 134, 60–67. [Google Scholar] [CrossRef]
  71. Ahmed, H.M.; Roy, A.; Wahab, M.; Ahmed, M.; Othman-Qadir, G.; Elesawy, B.H.; Khandaker, M.U.; Islam, M.N.; Emran, T.B. Applications of nanomaterials in agrifood and pharmaceutical industry. J. Nanomater. 2021, 2021, 1472096. [Google Scholar] [CrossRef]
  72. Brocchini, S.; Duncan, R. Pendent drugs release from polymers. In Encyclopedia of Controlled Drug Delivery, 1st ed.; John Wiley and Sons: New York, NY, USA, 1999; p. 561. [Google Scholar]
  73. Pillai, O.; Panchagnula, R. Polymers in drug delivery. Curr. Opin. Chem. Biol. 2001, 5, 447–451. [Google Scholar] [CrossRef]
  74. Maham, A.; Tang, Z.; Wu, H.; Wang, J.; Lin, Y. Protein-based nanomedicine platforms for drug delivery. Small 2009, 5, 1706–1721. [Google Scholar] [CrossRef]
  75. Cavalu, S.; Bisboaca, S.; Mates, I.M.; Pasca, P.M.; Laslo, V.; Costea, T.; Fritea, L.; Vicas, S. Novel Formulation Based on Chitosan-Arabic Gum Nanoparticles Entrapping Propolis Extract Production, physico-chemical and structural characterization. Rev. De Chim. 2018, 69, 3756–3760. [Google Scholar] [CrossRef]
  76. Avcu, E.; Bastan, F.E.; Guney, M.; Avcu, Y.Y.; Rehman, M.A.U.; Boccaccini, A.R. Biodegradable polymer matrix composites containing graphene-related materials for antibacterial applications: A critical review. Acta Biomater. 2022, in press. [CrossRef]
  77. Arif, Z.U.; Khalid, M.Y.; Noroozi, R.; Sadeghianmaryan, A.; Jalalvand, M.; Hossain, 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] [PubMed]
  78. Kenry, L.; Liu, B. Recent advances in biodegradable conducting polymers and their biomedical applications. Biomacromolecules 2018, 19, 1783–1803. [Google Scholar] [CrossRef] [PubMed]
  79. Rai, P.; Mehrotra, S.; Priya, S.; Gnansounou, E.; Sharma, S.K. Recent advances in the sustainable design and applications of biodegradable polymers. Bioresour. Technol. 2021, 325, 124739. [Google Scholar] [CrossRef] [PubMed]
  80. Lee, D.H.; Park, T.; Yoo, H. Biodegradable polymer composites for electrophysiological signal sensing. Polymers 2022, 14, 2875. [Google Scholar] [CrossRef] [PubMed]
  81. Ansari, M.J.; Rajendran, R.R.; Mohanto, S.; Agarwal, U.; Panda, K.; Dhotre, K.; Manne, R.; Deepak, A.; Zafar, A.; Yasir, M.; et al. Poly(N-isopropylacrylamide)-based hydrogels for biomedical applications: A review of the state-of-the-art. Gels 2022, 8, 454. [Google Scholar] [CrossRef]
  82. Mashak, A.; Rahimi, A. Silicone polymers in controlled drug delivery systems: A review. Iran. Polymer J. 2009, 18, 279–295. [Google Scholar]
  83. Priya James, H.; John, R.; Alex, A.; Anoop, K.R. Smart polymers for the controlled delivery of drugs—A concise overview. Acta Pharm. Sin. B 2014, 4, 120–127. [Google Scholar] [CrossRef]
  84. Teodorescu, M.; Bercea, M. Poly (vinylpyrrolidone)—A versatile polymer for biomedical and beyond medical applications. Polym. Plast. Technol. Eng. 2015, 54, 923–943. [Google Scholar] [CrossRef]
  85. Kaushik, R.; Budhwar, V.; Kaushik, D. An Overview on recent patents and technologies on solid dispersion. Recent Pat. Drug Deliv. Formul. 2020, 14, 63–74. [Google Scholar] [CrossRef]
  86. Karthika, C.; Hari, B.; Mano, V.; Radhakrishnan, A.; Janani, S.K.; Akter, R.; Kaushik, D.; Rahman, M.H. Curcumin as a great contributor for the treatment and mitigation of colorectal cancer. Exp. Gerontol. 2021, 152, 111438. [Google Scholar] [CrossRef]
  87. Chouhan, N.; Mittal, V.; Kaushik, D.; Khatkar, A.; Raina, M. Self emulsifying drug delivery system (SEDDS) for phytoconstituents: A review. Curr. Drug Deliv. 2015, 12, 244–253. [Google Scholar] [CrossRef] [PubMed]
  88. Kim, K.; Yoo, H.; Lee, E.K. New Opportunities for organic semiconducting polymers in biomedical applications. Polymers 2022, 14, 2960. [Google Scholar] [CrossRef]
  89. Win, K.Y.; Feng, S.S. Effects of particle size and surface coating on cellular uptake of polymeric nanoparticles for oral delivery of anticancer drugs. Biomaterials 2005, 26, 2713–2722. [Google Scholar] [CrossRef]
  90. He, C.; Yin, L.; Tang, C.; Yin, C. Size-dependent absorption mechanism of polymeric nanoparticles for oral delivery of protein drugs. Biomaterials 2012, 33, 8569–8578. [Google Scholar] [CrossRef] [PubMed]
  91. Kulkarni, S.A.; Feng, S.S. Effects of particle size and surface modification on cellular uptake and biodistribution of polymeric nanoparticles for drug delivery. Pharm Res. 2013, 30, 2512–2522. [Google Scholar] [CrossRef]
  92. Pandey, R.; Ahmad, Z.; Sharma, S.; Khuller, G.K. Nano-encapsulation of azole antifungals: Potential applications to improve oral drug delivery. Int. J. Pharm. 2005, 301, 268–276. [Google Scholar] [CrossRef] [PubMed]
  93. Ahlin, P.; Kristl, J.; Kristl, A.; Vrecer, F. Investigation of polymeric nanoparticles as carriers of enalaprilat for oral administration. Int. J. Pharm. 2002, 239, 113–120. [Google Scholar] [CrossRef]
  94. Pandey, R.; Zahoor, A.; Sharma, S.; Khuller, G.K. Nanoparticle encapsulated antitubercular drugs as a potential oral drug delivery system against murine tuberculosis. Tuberculosis 2003, 83, 373–378. [Google Scholar] [CrossRef]
  95. Roger, E.; Kalscheuer, S.; Kirtane, A.; Guru, B.R.; Grill, A.E.; Whittum-Hudson, J.; Panyam, J. Folic acid functionalized nanoparticles for enhanced oral drug delivery. Mol. Pharm. 2012, 9, 2103–2110. [Google Scholar] [CrossRef]
  96. Damgé, C.; Maincent, P.; Ubrich, N. Oral delivery of insulin associated to polymeric nanoparticles in diabetic rats. J. Contr. Release 2007, 117, 163–170. [Google Scholar] [CrossRef]
  97. Jain, A.K.; Swarnakar, N.K.; Das, M.; Godugu, C.; Singh, R.P.; Rao, P.R.; Jain, S. Augmented anticancer efficacy of doxorubicin-loaded polymeric nanoparticles after oral administration in a breast cancer induced animal model. Mol. Pharm. 2011, 8, 1140–1151. [Google Scholar] [CrossRef] [PubMed]
  98. Jana, S.; Maji, N.; Nayak, A.K.; Sen, K.K.; Basu, S.K. Development of chitosan-based nanoparticles through inter-polymeric complexation for oral drug delivery. Carbohydr. Polym. 2013, 98, 870–876. [Google Scholar] [CrossRef] [PubMed]
  99. He, C.; Yin, L.; Tang, C.; Yin, C. Multifunctional polymeric nanoparticles for oral delivery of TNF-α siRNA to macrophages. Biomaterials 2013, 34, 2843–2854. [Google Scholar] [CrossRef] [PubMed]
  100. Cavalu, S.; Antoniac, I.V.; Mohan, A.; Bodog, F.; Doicin, C.; Mates, I.; Ulmeanu, M.; Murzac, R.; Semenescu, A. Nanoparticles and Nanostructured Surface Fabrication for Innovative Cranial and Maxillofacial Surgery. Materials 2020, 13, 5391. [Google Scholar] [CrossRef] [PubMed]
  101. Cohen, H.; Levy, R.J.; Gao, J.; Fishbein, I.; Kousaev, V.; Sosnowski, S.; Slomkowski, S.; Golomb, G. Sustained delivery and expression of DNA encapsulated in polymeric nanoparticles. Gene Ther. 2000, 7, 1896–1905. [Google Scholar] [CrossRef]
  102. Jain, A.K.; Swarnakar, N.K.; Godugu, C.; Singh, R.P.; Jain, S. The effect of the oral administration of polymeric nanoparticles on the efficacy and toxicity of tamoxifen. Biomaterials 2011, 32, 503–515. [Google Scholar] [CrossRef]
  103. Malik, B.; Goyal, A.K.; Markandeywar, T.S.; Rath, G.; Zakir, F.; Vyas, S.P. Microfold-cell targeted surface engineered polymeric nanoparticles for oral immunization. J. Drug Target. 2012, 20, 76–84. [Google Scholar] [CrossRef]
  104. Zhang, Y.; Li, X.; Zhou, Y.; Wang, X.; Fan, Y.; Huang, Y.; Liu, Y. Preparation and evaluation of poly(ethylene glycol)-poly(lactide) micelles as nanocarriers for oral delivery of cyclosporine A. Nanoscale Res. Lett. 2010, 5, 917–925. [Google Scholar] [CrossRef]
  105. Ghosh, R.; Mondal, S.; Mukherjee, D.; Adhikari, A.; Ahmed, S.A.; Alsantali, R.I. Oral drug delivery using a polymeric nanocarrier: Chitosan nanoparticles in the delivery of rifampicin. Mater. Adv. 2022, 3, 4622–4628. [Google Scholar] [CrossRef]
  106. Akib, A.A.; Rumon, M.H.; Moniruzzaman, M.; Chowdhury, A.N.; Roy, C.K. PLA-PEG Diblock Copolymer Micelle as Nanocarrier for Anti-Obesity Drug Delivery Dystem. SPAST. 2021 Abstracts, 1(01). Available online: https://spast.org/techrep/article/view/1901 (accessed on 11 March 2022).
  107. Chen, T.; Tu, L.; Wang, G.; Qi, N.; Wu, W.; Zhang, W.; Feng, J. Multi-functional chitosan polymeric micelles as oral paclitaxel delivery systems for enhanced bioavailability and anti-tumor efficacy. Int. J. Pharm. 2020, 578, 119105. [Google Scholar] [CrossRef]
  108. Zhu, C.; Gong, S.; Ding, J.; Yu, M.; Ahmad, E.; Feng, Y.; Gan, Y. Supersaturated polymeric micelles for oral silybin delivery: The role of the Soluplus-PVPVA complex. Acta Pharm. Sin. B 2019, 9, 107–117. [Google Scholar] [CrossRef] [PubMed]
  109. Salimi, A.; Makhmal, S.; Zadeh, B.; Kazemi, M. Preparation and optimization of polymeric micelles as an oral drug delivery system for deferoxamine mesylate: In vitro and ex vivo studies. Res. Pharm. Sci. 2019, 14, 293–307. [Google Scholar] [CrossRef] [PubMed]
  110. Woraphatphadung, T.; Sajomsang, W.; Rojanarata, T. Development of chitosan-based pH-sensitive polymeric micelles containing curcumin for colon-targeted drug delivery. AAPS Pharm. Sci. Tech. 2018, 19, 991–1000. [Google Scholar] [CrossRef] [PubMed]
  111. Motwani, S.K.; Chopra, S.; Talegaonkar, S.; Kohli, K.; Ahmad, F.J.; Khar, R.K. Chitosan-sodium alginate nanoparticles as submicroscopic reservoirs for ocular delivery: Formulation, optimisation and in vitro characterisation. Eur. J. Pharm. Biopharm. 2008, 68, 513–525. [Google Scholar] [CrossRef]
  112. Kumari, A.; Yadav, S.K.; Yadav, S.C. Biodegradable polymeric nanoparticles based drug delivery systems. Colloids Surf. B Biointerfaces 2010, 75, 1–18. [Google Scholar] [CrossRef]
  113. Nagarwal, R.C.; Kant, S.; Singh, P.N.; Maiti, P.; Pandit, J.K. Polymeric nanoparticulate system: A potential approach for ocular drug delivery. J. Contr Releas. 2009, 136, 2–13. [Google Scholar] [CrossRef]
  114. Sharma, U.K.; Verma, A.; Prajapati, S.K.; Pandey, H.; Pandey, A.C. In vitro, in vivo and pharmacokinetic assessment of amikacin sulphate laden polymeric nanoparticles meant for controlled ocular drug delivery. Appl. Nanosci. 2014, 5, 143–155. [Google Scholar] [CrossRef]
  115. Mahor, A.; Prajapati, S.K.; Verma, A.; Gupta, R.; Iyer, A.K.; Kesharwani, P. Moxifloxacin loaded gelatin nanoparticles for ocular delivery: Formulation and in vitro, in vivo evaluation. J. Colloid Interface Sci. 2016, 483, 132–138. [Google Scholar] [CrossRef]
  116. Mittal, N.; Kaur, G. Investigations on polymeric nanoparticles for ocular delivery. Adv. Polym. 2019, 2019, 1316249. [Google Scholar] [CrossRef]
  117. Duxfield, L.; Sultana, R.; Wang, R.; Englebretsen, V.; Deo, S.; Swift, S.; Rupenthal, I.; Al-Kassas, R. Development of gatifloxacin-loaded cationic polymeric nanoparticles for ocular drug delivery. Pharm. Dev. Technol. 2016, 21, 172–179. [Google Scholar] [CrossRef]
  118. Musumeci, T.; Bucolo, C.; Carbone, C.; Pignatello, R.; Drago, F.; Puglisi, G. Polymeric nanoparticles augment the ocular hypotensive effect of melatonin in rabbits. Int. J. Pharm. 2013, 440, 135–140. [Google Scholar] [CrossRef]
  119. Costa, J.R.; Silva, N.C.; Sarmento, B.; Pintado, M. Potential chitosan-coated alginate nanoparticles for ocular delivery of daptomycin. Eur. J. Clin. Microbiol. Infect. Dis. 2015, 34, 1255–1262. [Google Scholar] [CrossRef] [PubMed]
  120. Aksungur, P.; Demirbilek, M.; Denkbaş, E.B.; Vandervoort, J.; Ludwig, A.; Unlü, N. Development and characterization of cyclosporine A loaded nanoparticles for ocular drug delivery: Cellular toxicity, uptake and kinetic studies. J. Control. Release 2011, 151, 286–294. [Google Scholar] [CrossRef] [PubMed]
  121. Yoncheva, K.; Vandervoort, J.; Ludwig, A. Development of mucoadhesive poly(lactide-co-glycolide) nanoparticles for ocular application. Pharm. Dev. Technol. 2011, 16, 29–35. [Google Scholar] [CrossRef]
  122. Gadad, A.P.; Chandra, P.S.; Dandagi, P.M.; Mastiholimath, V.S. Moxifloxacin loaded polymeric nanoparticles for sustained ocular drug delivery. Int. J. Pharm. Sci. Nanotechnol. 2012, 5, 1727–1734. [Google Scholar] [CrossRef]
  123. Singh, K.H.; Shinde, U.A. Development and evaluation of novel polymeric nanoparticles of brimonidine tartrate. Curr. Drug Deliv. 2010, 7, 244–251. [Google Scholar] [CrossRef]
  124. Jain, K.; Kumar, R.S.; Sood, S.; Dhyanandhan, G. Betaxolol hydrochloride loaded chitosan nanoparticles for ocular delivery and their anti-glaucoma efficacy. Curr. Drug Deliv. 2013, 10, 493–499. [Google Scholar] [CrossRef]
  125. Başaran, E.; Demirel, M.; Sirmagül, B.; Yazan, Y. Polymeric cyclosporine-A nanoparticles for ocular application. J. Biomed. Nanotechnol. 2011, 7, 714–723. [Google Scholar] [CrossRef]
  126. Nasr, F.H.; Khoee, S.; Dehghan, M.M.; Chaleshtori, S.S.; Shafiee, A. Preparation and evaluation of contact lenses embedded with polycaprolactone-based nanoparticles for ocular drug delivery. Biomacromolecules 2016, 17, 485–495. [Google Scholar] [CrossRef]
  127. Shmueli, R.B.; Sunshine, J.C.; Xu, Z.; Duh, E.J.; Green, J.J. Gene delivery nanoparticles specific for human microvasculature and macrovasculature. Nanomedicine 2012, 8, 1200–1207. [Google Scholar] [CrossRef]
  128. Sah, A.K.; Suresh, P.K.; Verma, V.K. PLGA nanoparticles for ocular delivery of loteprednol etabonate: A corneal penetration study. Artif. Cells Nanomed. Biotechnol. 2017, 45, 1156–1164. [Google Scholar] [CrossRef] [PubMed]
  129. Basaran, E.; Senel, B.; Kirimlioglu, G.Y.; Guven, U.M.; Yazan, Y. Ornidazole incorporated chitosan nanoparticles for ocular application. Latin Am. J. Pharm. 2015, 34, 1180–1188. [Google Scholar]
  130. Zhang, P.; Liu, X.; Hu, W.; Bai, Y.; Zhang, L. Preparation and evaluation of naringenin-loaded sulfobutylether-β-cyclodextrin/chitosan nanoparticles for ocular drug delivery. Carbohydr. Polym. 2016, 149, 224–230. [Google Scholar] [CrossRef] [PubMed]
  131. Xu, L.; Xu, X.; Chen, H.; Li, X. Ocular biocompatibility and tolerance study of biodegradable polymeric micelles in the rabbit eye. Colloids Surf. B Biointerfaces 2013, 112, 30–34. [Google Scholar] [CrossRef] [PubMed]
  132. 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]
  133. Gupta, H.; Aqil, M.; Khar, R.K.; Ali, A.; Bhatnagar, A.; Mittal, G. Sparfloxacin-loaded PLGA nanoparticles for sustained ocular drug delivery. Nanomedicine 2010, 6, 324–333. [Google Scholar] [CrossRef]
  134. Salama, A.H.; Shamma, R.N. Tri/tetra-block co-polymeric nanocarriers as a potential ocular delivery system of lornoxicam: In vitro characterization.; and in vivo estimation of corneal permeation. Int. J. Pharm. 2015, 492, 28–39. [Google Scholar] [CrossRef]
  135. Pignatello, R.; Ricupero, N.; Bucolo, C.; Maugeri, F.; Maltese, A.; Puglisi, G. Preparation and characterization of Eudragit retard nanosuspensions for the ocular delivery of cloricromene. AAPS Pharm. Sci. Tech. 2006, 7, 192–198. [Google Scholar] [CrossRef]
  136. Vega, E.; Gamisans, F.; García, M.L.; Chauvet, A.; Lacoulonche, F.; Egea, M.A. PLGA nanospheres for the ocular delivery of flurbiprofen: Drug release and interactions. J. Pharm. Sci. 2008, 97, 5306–5317. [Google Scholar] [CrossRef]
  137. Katara, R.; Majumdar, D.K. Eudragit RL 100-based nanoparticulate system of aceclofenac for ocular delivery. Colloids Surf. B Biointerfaces 2013, 103, 455–462. [Google Scholar] [CrossRef]
  138. Salama, A.H.; Mahmoud, A.A.; Kamel, R. A novel method for preparing surface-modified fluocinolone acetonide loaded PLGA nanoparticles for ocular use: In vitro and in vivo evaluations. AAPS Pharm. Sci. Tech. 2016, 17, 1159–1172. [Google Scholar] [CrossRef] [PubMed]
  139. Khin, S.Y.; Soe, H.M.S.H.; Chansriniyom, C.; Pornputtapong, N.; Asasutjarit, R.; Loftsson, T.; Jansook, P. Development of fenofibrate/randomly methylated β-cyclodextrin-loaded Eudragit® RL 100 nanoparticles for ocular delivery. Molecules 2022, 27, 4755. [Google Scholar] [CrossRef]
  140. Jacinto, T.A.; Oliveira, B.; Miguel, S.P.; Ribeiro, M.P.; Coutinho, P. Ciprofloxacin-loaded zein/hyaluronic acid nanoparticles for ocular mucosa delivery. Pharmaceutics 2022, 14, 1557. [Google Scholar] [CrossRef] [PubMed]
  141. Mohamed, H.B.; Attia Shafie, M.A.; Mekkawy, A.I. Chitosan nanoparticles for meloxicam ocular delivery: Development, in vitro characterization, and in vivo evaluation in a rabbit eye model. Pharmaceutics 2022, 14, 893. [Google Scholar] [CrossRef] [PubMed]
  142. Prosperi-Porta, G.; Kedzior, S.; Muirhead, B.; Sheardown, H. Phenylboronic-acid-based polymeric micelles for mucoadhesive anterior segment ocular drug delivery. Biomacromolecules 2016, 17, 1449–1457. [Google Scholar] [CrossRef]
  143. Pignatello, R.; Corsaro, R.; Bonaccorso, A.; Zingale, E.; Carbone, C.; Musumeci, T. Soluplus® polymeric nanomicelles improve solubility of BCS-class II drugs. Drug Deliv. Transl. Res. 2022, 12, 1991–2006. [Google Scholar] [CrossRef]
  144. Binkhathlan, Z.; Alomrani, A.H.; Hoxha, O.; Ali, R.; Kalam, M.A.; Alshamsan, A. Development and characterization of PEGylated fatty acid-block-poly(ε-caprolactone) novel block copolymers and their self-assembled nanostructures for ocular delivery of cyclosporine A. Polymers 2022, 14, 1635. [Google Scholar] [CrossRef]
  145. Safwat, M.A.; Mansour, H.F.; Hussein, A.K.; Abdelwahab, S.; Soliman, G.M. Polymeric micelles for the ocular delivery of triamcinolone acetonide: Preparation and in vivo evaluation in a rabbit ocular inflammatory model. Drug Deliv. 2020, 27, 1115–1124. [Google Scholar] [CrossRef]
  146. Varela-Fernández, R.; García-Otero, X.; Díaz-Tomé, V.; Regueiro, U.; López-López, M.; González-Barcia, M.; Isabel Lema, M.; Otero-Espinar, F.J. Mucoadhesive PLGA nanospheres and nanocapsules for lactoferrin controlled ocular delivery. Pharmaceutics 2022, 14, 799. [Google Scholar] [CrossRef]
  147. Silva-Abreu, M.; Calpena, A.C.; Espina, M. Optimization, biopharmaceutical profile and therapeutic efficacy of pioglitazone-loaded PLGA-PEG nanospheres as a novel strategy for ocular inflammatory disorders. Pharm Res. 2018, 35, 11. [Google Scholar] [CrossRef]
  148. Muthu, M.S. Nanoparticles based on PLGA and its co-polymer: An overview. Asian J. Pharm. Sci. 2009, 3, 266–273. [Google Scholar] [CrossRef]
  149. Ulbrich, K.; Holá, K.; Šubr, V.; Bakandritsos, A.; Tuček, J.; Zbořil, R. Targeted drug delivery with polymers and magnetic nanoparticles: Covalent and noncovalent approaches, release control, and clinical studies. Chem. Rev. 2016, 116, 5338–5431. Available online: https://pubs.acs.org/doi/full/10.1021/acs.chemrev.5b00589 (accessed on 4 March 2022). [CrossRef] [PubMed]
  150. Tosi, G.; Bortot, B.; Ruozi, B.; Dolcetta, D.; Vandelli, M.A.; Forni, F.; Severini, G.M. Potential use of polymeric nanoparticles for drug delivery across the blood-brain barrier. Curr. Med. Chem. 2013, 20, 2212–2225. [Google Scholar] [CrossRef]
  151. Muthu, M.S.; Singh, S. Studies on biodegradable polymeric nanoparticles of risperidone: In vitro and in vivo evaluation. Nanomedicine 2008, 3, 305–319. [Google Scholar] [CrossRef]
  152. Muthu, M.S.; Rawat, M.K.; Mishra, A.; Singh, S. PLGA nanoparticle formulations of risperidone: Preparation and neuropharmacological evaluation. Nanomedicine 2009, 5, 323–333. [Google Scholar] [CrossRef] [PubMed]
  153. Bisht, S.; Mizuma, M.; Feldmann, G.; Ottenhof, N.A.; Hong, S.M.; Pramanik, D.; Chenna, V.; Karikari, C.; Sharma, R.; Goggins, M.G.; et al. Systemic administration of polymeric nanoparticle-encapsulated curcumin (NanoCurc) blocks tumor growth and metastases in preclinical models of pancreatic cancer. Mol. Cancer Ther. 2010, 9, 2255–2264. [Google Scholar] [CrossRef] [PubMed]
  154. Nanjwade, B.K.; Singh, J.; Parikh, K.A.; Manvi, F.V. Preparation and evaluation of carboplatin biodegradable polymeric nanoparticles. Int. J. Pharm. 2010, 385, 176–180. [Google Scholar] [CrossRef]
  155. Song, L.; Shen, Y.; Hou, J.; Lei, L.; Guo, S.; Qian, C. Polymeric micelles for parenteral delivery of curcumin: Preparation.; characterization and in vitro evaluation. Colloids Surf. 2011, 390, 25–32. [Google Scholar] [CrossRef]
  156. Richter, A.; Olbrich, C.; Krause, M.; Hoffmann, J.; Kissel, T. Polymeric micelles for parenteral delivery of sagopilone: Physicochemical characterization, novel formulation approaches and their toxicity assessment in vitro as well as in vivo. Eur. J. Pharm. Biopharm. 2010, 75, 80–89. [Google Scholar] [CrossRef]
  157. Haggag, Y.A.; Abosalha, A.K.; Tambuwala, M.M.; Silva, A.M.; Gimeno, A.; Egea, M.A.; García, M.L. Polymeric nanoencapsulation of zaleplon into PLGA nanoparticles for enhanced pharmacokinetics and pharmacological activity. Biopharm. Drug Dispos. 2021, 42, 12–23. [Google Scholar] [CrossRef]
  158. Ford, C.A.; Spoonmore, T.J.; Gupta, M.K.; Duvall, C.L.; Guelcher, S.A.; Cassat, J.E. Diflunisal-loaded poly(propylene sulfide) nanoparticles decrease S. aureus-mediated bone destruction during osteomyelitis. J. Orthop. Res. 2021, 39, 426–437. [Google Scholar] [CrossRef] [PubMed]
  159. Elmowafy, M.; Alruwaili, N.K.; Shalaby, K.; Alharbi, K.S.; Altowayan, W.M.; Ahmad, N.; Zafar, A.; Elkomy, M. Long-acting paliperidone parenteral formulations based on polycaprolactone nanoparticles; the influence of stabilizer and chitosan on in vitro release, protein adsorption, and cytotoxicity. Pharmaceutics 2020, 12, 160. [Google Scholar] [CrossRef] [PubMed]
  160. Sang, M.M.; Liu, F.L.; Wang, Y.; Luo, R.J.; Huan, X.X.; Han, L.F.; Zhang, Z.T.; Feng, F.; Qu, W.; Liu, W.; et al. A novel redox/pH dual-responsive and hyaluronic acid-decorated multifunctional magnetic complex micelle for targeted gambogic acid delivery for the treatment of triple negative breast cancer. Drug Deliv. 2018, 25, 1846–1857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Wang, Z.; Sau, S.; Alsaab, H.O.; Iyer, A.K. CD44 directed nanomicellar payload delivery platform for selective anticancer effect and tumor specific imaging of triple negative breast cancer. Nanomedicine 2018, 14, 1441–1454. [Google Scholar] [CrossRef] [PubMed]
  162. Hiroshi, M. Macromolecular therapeutics in cancer treatment: The EPR effect and beyond. J. Control. Release 2012, 164, 138–144. [Google Scholar] [CrossRef]
  163. Akhtar, N.; Verma, A.; Pathak, K. Topical delivery of drugs for the effective treatment of fungal infections of skin. Curr. Pharm. Des. 2015, 21, 2892–2913. [Google Scholar] [CrossRef]
  164. Akhtar, N.; Verma, A.; Pathak, K. Exploring preclinical and clinical effectiveness of nanoformulations in the treatment of atopic dermatitis: Safety aspects and patent reviews. Bull. Fac. Pharm. Cairo Univ. 2017, 55, 3. [Google Scholar] [CrossRef]
  165. Kim, D.G.; Jeong, Y.I.; Choi, C.; Roh, S.H.; Kang, S.K.; Jang, M.K.; Nah, J.W. Retinol-encapsulated low molecular water-soluble chitosan nanoparticles. Int. J. Pharm. 2006, 319, 130–138. [Google Scholar] [CrossRef]
  166. Elmowafy, M.; Samy, A.; Raslan, M.A.; Salama, A.; Said, R.A.; Abdelaziz, A.E.; El-Eraky, W.; El Awdan, S.; Viitala, T. Enhancement of bioavailability and pharmacodynamic effects of thymoquinone via nanostructured lipid carrier (NLC) formulation. AAPS Pharm. Sci. Tech. 2016, 17, 663–672. [Google Scholar] [CrossRef]
  167. Chen, Y.S.; Alany, R.G.; Young, S.A.; Green, C.R.; Rupenthal, I.D. In vitro release characteristics and cellular uptake of poly(D.;L-lactic-co-glycolic acid) nanoparticles for topical delivery of antisense oligodeoxynucleotides. Drug Deliv. 2011, 18, 493–501. [Google Scholar] [CrossRef]
  168. Youngren, S.R.; Tekade, R.K.; Gustilo, B.; Hoffmann, P.R.; Chougule, M.B. STAT6 siRNA matrix-loaded gelatin nanocarriers: Formulation, characterization, and ex vivo proof of concept using adenocarcinoma cells. Biomed. Res. Int. 2013, 2013, 858946. [Google Scholar] [CrossRef] [PubMed]
  169. Balzus, B.; Sahle, F.F.; Hönzke, S.; Gerecke, C.; Schumacher, F.; Hedtrich, S.; Kleuser, B.; Bodmeier, R. Formulation and ex vivo evaluation of polymeric nanoparticles for controlled delivery of corticosteroids to the skin and the corneal epithelium. Eur. J. Pharm. Biopharm. 2017, 115, 122–130. [Google Scholar] [CrossRef] [PubMed]
  170. Saadallah, M.A.; Hamid, O. Formulation and evaluation of rosuvastatin calcium polymeric nanoparticles-loaded transdermal patch. Iraqi J. Pharm. 2022, 18, 22–38. [Google Scholar] [CrossRef]
  171. Albanese, A.; Tang, P.S.; Chan, W.C. The effect of nanoparticle size, shape, and surface chemistry on biological systems. Annu. Rev. Biomed. Eng. 2012, 14, 1–16. [Google Scholar] [CrossRef] [PubMed]
  172. Kamaly, N.; Xiao, Z.; Valencia, P.M.; Radovic-Moreno, A.F.; Farokhzad, O.C. Targeted polymeric therapeutic nanoparticles: Design.; development and clinical translation. Chem. Soc. Rev. 2012, 41, 2971–3010. [Google Scholar] [CrossRef]
  173. Etheridge, M.L.; Campbell, S.A.; Erdman, A.G.; Haynes, C.L.; Wolf, S.M.; McCullough, J. The big picture on nanomedicine: The state of investigational and approved nanomedicine products. Nanomedicine 2013, 9, 1–14. [Google Scholar] [CrossRef]
  174. Cavalu, S.; Ratiu, C.; Ponta, O.; Simon, V.; Rugina, D.; Miclaus, V.; Akin, I.; Goller, G. Improving osseointegration of alumina/zirconia ceramic implants by fluoride surface treatment. Dig. J. Nanomater. Biostructures 2014, 9, 797–808. [Google Scholar]
  175. Bobo, D.; Robinson, K.J.; Islam, J.; Thurecht, K.J.; Corrie, S.R. Nanoparticle-based medicines: A review of FDA-approved materials and clinical trials to date. Pharm. Res. 2016, 33, 2373–2387. [Google Scholar] [CrossRef]
  176. Duncan, R. Polymer therapeutics: Top 10 selling pharmaceuticals—What next? J. Control. Release 2014, 190, 371–380. [Google Scholar] [CrossRef]
  177. Lang, L. FDA approves Cimzia to treat Crohn’s disease. Gastroenterology 2008, 134, 1819. [Google Scholar] [CrossRef]
  178. Krystexxa Pegloticase. 2020. Available online: https://www.krystexxa.com/ (accessed on 6 March 2020).
  179. Kolb-Mäurer, A.; Sunderkötter, C.; Kukowski, B.; Meuth, S.G. An update on peginterferon beta-1a management in multiple sclerosis: Results from an interdisciplinary board of German and Austrian neurologists and dermatologists. BMC Neurol. 2019, 19, 130. [Google Scholar] [CrossRef] [PubMed]
  180. Prime, B.I. CHMP Recommends EU Marketing Authorization for ADYNOVI® [Antihemophilic Factor (Recombinant), PEGylated] for Adults and Adolescents with Hemophilia, A. 2020. Available online: https://www.globenewswire.com/news-release/2017/11/13/1184850/0/en/CHMP-recommends-EU-marketing-authorization-for-ADYNOVI-Antihemophilic-Factor-Recombinant-PEGylated-for-adults-and-adolescents-with-Hemophilia-A.html (accessed on 8 July 2020).
  181. Piedmonte, D.M.; Treuheit, M.J. Formulation of Neulasta. Adv. Drug Deliv. Rev. 2008, 60, 50–58. [Google Scholar] [CrossRef] [PubMed]
  182. Rajender Reddy, K.; Modi, M.W.; Pedder, S. Use of peginterferon alfa-2a (40 KD) (Pegasys) for the treatment of hepatitis C. Adv. Drug Deliv. Rev. 2002, 54, 571–586. [Google Scholar] [CrossRef]
  183. Ge, Y.; Grossman, R.I.; Udupa, J.K.; Fulton, J.; Constantinescu, C.S.; Gonzales–Scarano, F.; Babb, J.S.; Mannon, L.J.; Kolson, D.L.; Cohen, J.A. Glatiramer acetate (Copaxone) treatment in relapsing-remitting MS: Quantitative MR assessment. Neurology 2000, 54, 813–817. [Google Scholar] [CrossRef]
  184. Oerlemans, C.; Bult, W.; Bos, M.; Storm, G.; Nijsen, J.F.; Hennink, W.E. Polymeric micelles in anticancer therapy: Targeting, imaging and triggered release. Pharm. Res. 2010, 27, 2569–2589. [Google Scholar] [CrossRef]
  185. Chiechi, L.M. Estrasorb. I Drugs 2004, 7, 860–864. [Google Scholar]
  186. U.S. National Library of Medicine. 2020. Available online: https://clinicaltrials.gov/ (accessed on 8 July 2020).
  187. Unites States Patent and Trademark Office. 2020. Available online: https://www.uspto.gov/ (accessed on 8 July 2020).
  188. Sharma, N.; Madan, P.; Lin, S. Effect of process and formulation variables on the preparation of parenteral paclitaxel-loaded biodegradable polymeric nanoparticles: A co-surfactant study. Asian J. Pharm. Sci. 2016, 11, 404–416. [Google Scholar] [CrossRef]
  189. Ahlawat, J.; Henriquez, G.; Narayan, M. Enhancing the delivery of chemotherapeutics: Role of biodegradable polymeric nanoparticles. Molecules 2018, 23, 2157. [Google Scholar] [CrossRef]
  190. Caster, J.M.; Stephanie, K.Y.; Patel, A.N.; Newman, N.J.; Lee, Z.J.; Warner, S.B.; Wagner, K.T.; Roche, K.C.; Tian, X.; Min, Y.; et al. Effect of particle size on the biodistribution, toxicity, and efficacy of drug-loaded polymeric nanoparticles in chemoradiotherapy. Nanomedicine 2017, 13, 1673–1683. [Google Scholar] [CrossRef]
  191. Singh, R.P.; Ramarao, P. Accumulated polymer degradation products as effector molecules in cytotoxicity of polymeric nanoparticles. Toxicol. Sci. 2013, 136, 131–143. [Google Scholar] [CrossRef]
  192. Grabowski, N.; Hillaireau, H.; Vergnaud, J.; Tsapis, N.; Pallardy, M.; Kerdine-Römer, S.; Fattal, E. Surface coating mediates the toxicity of polymeric nanoparticles towards human-like macrophages. Int. J. Pharm. 2015, 482, 75–83. [Google Scholar] [CrossRef] [PubMed]
  193. Rejinold, N.S.; Muthunarayanan, M.; Divyarani, V.V.; Sreerekha, P.R.; Chennazhi, K.P.; Nair, S.V.; Tamura, H.; Jayakumar, R. Curcumin-loaded biocompatible thermoresponsive polymeric nanoparticles for cancer drug delivery. J. Colloid Interface Sci. 2011, 360, 39–51. [Google Scholar] [CrossRef] [PubMed]
  194. Dandekar, P.; Dhumal, R.; Jain, R.; Tiwari, D.; Vanage, G.; Patravale, V. Toxicological evaluation of pH-sensitive nanoparticles of curcumin: Acute.; sub-acute and genotoxicity studies. Food Chem. Toxicol. 2010, 48, 2073–2089. [Google Scholar] [CrossRef] [PubMed]
  195. Karabasz, A.; Bzowska, M.; Szczepanowicz, K. Biomedical applications of multifunctional polymeric nanocarriers: A review of current literature. Int. J. Nanomed. 2020, 15, 8673–8696. [Google Scholar] [CrossRef]
  196. Homayun, B.; Lin, X.; Choi, H.J. Challenges and recent progress in oral drug delivery systems for Biopharmaceuticals. Pharmaceutics 2019, 11, 129. [Google Scholar] [CrossRef] [Green Version]
  197. Nabi, B.; Rehman, S.; Baboota, S.; Ali, J. Insights on oral drug delivery of lipid nanocarriers: A win-win solution for augmenting bioavailability of antiretroviral drugs. AAPS Pharm. Sci. Tech. 2019, 20, 60. [Google Scholar] [CrossRef]
  198. Alqahtani, M.S.; Kazi, M.; Alsenaidy, M.A.; Ahmad, M.Z. Advances in oral drug delivery. Front. Pharmacol. 2021, 12, 618411. [Google Scholar] [CrossRef]
  199. Mangla, B.; Javed, S.; Sultan, M.H.; Ahsan, W.; Aggarwal, G.; Kohli, K. Nanocarriers-assisted needle-free vaccine delivery through oral and intranasal transmucosal routes: A novel therapeutic conduit. Front. Pharmacol. 2022, 12, 757761. [Google Scholar] [CrossRef]
  200. Tsai, C.H.; Wang, P.Y.; Lin, I.C.; Huang, H.; Liu, G.S.; Tseng, C.L. Ocular drug delivery: Role of degradable polymeric nanocarriers for ophthalmic application. Int. J. Mol. Sci. 2018, 19, 2830. [Google Scholar] [CrossRef]
  201. Grund, S.; Bauer, M.; Fischer, D. Polymers in drug delivery-state of the art and future trends. Adv. Eng. Mater. 2011, 13, 61–87. [Google Scholar] [CrossRef]
  202. Bennet, D.; Kim, S. Polymer nanoparticles for smart drug delivery. In Application of Nanotechnology in Drug Delivery; Sezer, A.D., Ed.; Intech: Rijeka, Croatia, 2014. [Google Scholar] [CrossRef]
  203. Luk, B.T.; Zhang, L. Current advances in polymer-based nanotheranostics for cancer treatment and diagnosis. ACS Appl. Mater. Interfaces 2014, 6, 21859–21873. [Google Scholar] [CrossRef] [PubMed]
  204. Vaut, L.; Juszczyk, J.J.; Kamguyan, K.; Jensen, K.E.; Tosello, G.; Boisen, A. 3D printing of reservoir devices for oral drug delivery: From concept to functionality through design improvement for enhanced mucoadhesion. ACS Biomater. Sci. Eng. 2020, 6, 2478–2486. [Google Scholar] [CrossRef] [PubMed]
  205. Vauthier, C.; Bouchemal, K. Processing and scale-up of polymeric nanoparticles. In Intracellular Delivery; Springer: Berlin/Heidelberg, Germany, 2011; pp. 433–456. [Google Scholar]
  206. Liechty, W.B.; Kryscio, D.R.; Slaughter, B.V.; Peppas, N.A. Polymers for drug delivery systems. Annu. Rev. Chem. Biomol. Eng. 2010, 1, 149–173. [Google Scholar] [CrossRef]
  207. Narang, A.S.; Chang, R.K.; Hussain, M.A. Pharmaceutical development and regulatory considerations for nanoparticles and nanoparticulate drug delivery systems. J. Pharm. Sci. 2013, 102, 3867–3882. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Diagrammatic representation of different polymeric nanocarriers.
Figure 1. Diagrammatic representation of different polymeric nanocarriers.
Polymers 14 03545 g001
Figure 2. Illustration of (A) TEM analysis; (B) in vitro drug release studies; (C) in vivo antitumor efficacy studies on B16F10 melanoma bearing mice; (D) Results of in vitro cellular uptake studies of C6 loaded FA-targeted PF127 Chol micelles, C6 loaded non-targeted PF127 Chol micelles, free C6, and culture media (Control) via (a) fluorescent microscopy and (b) visible light microscopy. Reproduced from Varshosaz et al., 2015 [35] under creative commons CC BY license © 2015, The Author(s).
Figure 2. Illustration of (A) TEM analysis; (B) in vitro drug release studies; (C) in vivo antitumor efficacy studies on B16F10 melanoma bearing mice; (D) Results of in vitro cellular uptake studies of C6 loaded FA-targeted PF127 Chol micelles, C6 loaded non-targeted PF127 Chol micelles, free C6, and culture media (Control) via (a) fluorescent microscopy and (b) visible light microscopy. Reproduced from Varshosaz et al., 2015 [35] under creative commons CC BY license © 2015, The Author(s).
Polymers 14 03545 g002
Figure 3. Illustration of (1) morphological analysis of (A) blank cubosomes, (B) uncoated CIS, (C) uncoated PAX, (D) uncoated DUAL, (E) coated CIS, (F) coated PAX and (G) coated DUAL cubosomes; (2) in vitro drug release studies (A) assembly and (B) comparative results of drug release; and (3) Cell viability studies on exposure of Human hepatoma HepG2 cells to (A) PBS blank cubosomes, (B) control cubosomes, (C) coated CIS, (D) uncoated CIS, (E) coated PAX, (F) uncoated PAX and (G) coated DUAL, (H) uncoated DUAL cubosomes. Reproduced from Zhang et al., 2020 [69] under creative commons CC BY license © 2020, The Author(s).
Figure 3. Illustration of (1) morphological analysis of (A) blank cubosomes, (B) uncoated CIS, (C) uncoated PAX, (D) uncoated DUAL, (E) coated CIS, (F) coated PAX and (G) coated DUAL cubosomes; (2) in vitro drug release studies (A) assembly and (B) comparative results of drug release; and (3) Cell viability studies on exposure of Human hepatoma HepG2 cells to (A) PBS blank cubosomes, (B) control cubosomes, (C) coated CIS, (D) uncoated CIS, (E) coated PAX, (F) uncoated PAX and (G) coated DUAL, (H) uncoated DUAL cubosomes. Reproduced from Zhang et al., 2020 [69] under creative commons CC BY license © 2020, The Author(s).
Polymers 14 03545 g003
Figure 4. Illustration of mucoadhesion property of polymeric nanoparticles (right) and micelles (left) in gastrointestinal transit time modulation resulting in enhanced mucosal penetration and oral bioavailability. ① receptor-mediated endocytosis, ② transcellular transport, ③ paracellular transport, and ④ M cell-mediated transport.
Figure 4. Illustration of mucoadhesion property of polymeric nanoparticles (right) and micelles (left) in gastrointestinal transit time modulation resulting in enhanced mucosal penetration and oral bioavailability. ① receptor-mediated endocytosis, ② transcellular transport, ③ paracellular transport, and ④ M cell-mediated transport.
Polymers 14 03545 g004
Figure 5. Illustration of ionic interaction of polymeric nanocarriers with corneal glycocalyx domain resulting in increased ocular surface contact and improved drug penetration.
Figure 5. Illustration of ionic interaction of polymeric nanocarriers with corneal glycocalyx domain resulting in increased ocular surface contact and improved drug penetration.
Polymers 14 03545 g005
Figure 6. Graphical representation of active and passive targeting of drugs via polymeric nanocarriers following parenteral administration.
Figure 6. Graphical representation of active and passive targeting of drugs via polymeric nanocarriers following parenteral administration.
Polymers 14 03545 g006
Table 1. Applications of nanopolymers in pharmaceutical and clinical medicine.
Table 1. Applications of nanopolymers in pharmaceutical and clinical medicine.
ClassificationName of PolymersApplications of PolymerRef.
Natural polymers
Protein basedSoy, whey, collagen, gelatinGene delivery, nanoparticles[74]
PolysaccharidesAlginate, pectin, guar gum, chitosan, chondroitin, heparin, hyaluronic acid, cyclodextrinsBinding and film coating agents in tablets, mucoadhesive, controlled release of drugs[75]
Synthetic polymers
Biodegradable
PolyamidesPolyamino acids, poly(iminocarbonates) and their copolymersSutures, catheters for angioplasty, controlled and sustained drug delivery[76]
CelluloseCarboxy methylcellulose, ethyl cellulose, cellulose acetate, HPMCBinder, coating, emulsifying, disintegrants in tablets and capsules[77]
PolyanhydridesPoly(adipic) acid, poly(sebacic) acid, and their copolymersControlled release coatings, medical implants[78]
PolyestersPoly (lactic acid), poly (glycolic acid), poly(dioxanes) and their copolymersProtein delivery, dialysis membrane[79]
OthersPolyurethane, polyortho esters, poly(cyano) acrylates, polystyrenesSutures, stents, drug delivery devices, dialysis media
Non-biodegradable
SiliconesColloidal silica, polydimethylsiloxaneTherapeutic devices, implants, medical grade adhesive for transdermal delivery[82]
Acrylic polymersPolymethacrylates, polyhydroxy (ethyl acrylates)Thermo-gelling acrylamide derivatives, its balance of hydrogen bonding, and hydrophobic association changes with temperature (smart polymers), film-forming agent[83]
OthersPVP, poloxamers, ethyl vinyl acetateTablet granulation, plasma replacement[84]
Table 2. Various nanopolymers employed as nanocarriers for oral delivery of drugs, peptides, and genes.
Table 2. Various nanopolymers employed as nanocarriers for oral delivery of drugs, peptides, and genes.
PolymerDelivery SystemTherapeutic MoleculeSummaryRef.
PLGANPsCoumarin-6Surface modification of PLGA nanoparticles with vitamin E TPGS notably improved the cellular uptake to promote oral chemotherapy.[89]
Carboxylated chitosanNPsBovine serum albuminChitosan grafted nanoparticles showed increased intestinal absorption due to higher mucoadhesion, cellular uptake, and systemic biodistribution after oral administration.[90]
PolystyreneNPsNAVitamin E TPGS modified polystyrene nanoparticles evidenced increased cellular uptake by Caco-2 and MDCK cells employed as an in vitro model for gastro-intestinal and blood-brain barrier respectively. In vivo investigation displayed that vitamin E TPGS coated nanoparticles (<200 nm) can escape RES and thus enhance half-life.[91]
PLGA and alginate stabilized chitosanNPsClotrimazole and econazoleOral administration of polymeric nanoparticle encapsulated drugs showed controlled drug delivery for 5–6 days compared to unencapsulated drugs which were cleared within 3–4 h following oral or IV administration. Bioavailability of drugs was remarkably improved and was detected in the lung, liver, and spleen tissues till 6–8 days compared to free drugs which were cleared by 12 h.[92]
PLGA and polymethylmethacrylateNPsEnalaprilatThe in vitro study across rat jejunum showed that the apparent permeability coefficient of enalaprilat-loaded PLGA nanoparticles was not significantly improved compared to the enalaprilat solution.[93]
PLGNPsRifampicin, isoniazid and pyrazinamideOral administration of drug-loaded nanoparticles in mice showed prolonged blood circulation for up to 6 days for rifampicin and up to 9 days for isoniazid and pyrazinamide. The therapeutic concentration of the drug in the tissues was maintained for 9–11 days.[94]
N-isopropyl acrylamide, methylmethacrylate and acrylic acid in 60:20:20NPsRapamycinSignificant blood levels of rapamycin were observed within 30 min after oral administration of rapamycin-loaded polymeric nanoparticles and continue to be detected in bloodstream up to 24 h.[88]
PLGANPsPaclitaxelOrally administered paclitaxel-loaded PLGA nanoparticles displayed a 5-fold increase in apparent permeability across Caco-2 cells, employed as in vitro model compared to free paclitaxel. Surface functionalization with folic acid further increased the transport of nanoparticles by 8-fold.[95]
Eudragit® RSNPsInsulinOral delivery of polymeric nanoparticles preserved the biological activity of encapsulated insulin and showed increased serum insulin level for a prolonged period due to the mucoadhesive property of polycationic polymer (Eudragit® RS) facilitating insulin intestinal uptake.[96]
PLGANPsDoxorubicinTime and concentration-dependent increase in cellular uptake of doxorubicin-loaded polymeric nanoparticles was observed across Caco-2 cells compared to doxorubicin solution. Orally administered nanoparticles displayed reduced cardiotoxicity compared with the intravenously injected free drug solution.[97]
ChitosanNPsAlprazolamThe cationic chitosan polymer was intercomplexed with anionic egg albumin and stabilized with PEG 400 to develop nanoparticles which demonstrated sustained drug delivery up to 24 h. [98]
Trimethyl chitosanNPsTNF-α siRNAFollowing oral administration mannose functionalized chitosan-cysteine conjugate nanoparticles enhanced siRNA stability in physiological fluid and promoted its transport across intestinal epithelium leading to siRNA uptake by macrophages through endocytosis and cytoplasmic siRNA release.[99]
PCL, PLGA and Eudrajit® RS and RLNPsHeparinOrally administered polymeric nano-particulate heparin showed prolonged anti-Xa activity compared to the heparin solution injected intravenously. A 2-fold increase in activated partial thromboplastin time was reported.[100]
Poly-(DL-lactide-co-glycolide)NPspDNA (alkaline phosphatase, a reporter gene)pDNA loaded in PLGA polymer particles showed sustained release of pDNA while maintaining its structural and functional integrity for over a month.[101]
PLGANPsTamoxifenThe oral bioavailability of tamoxifen was increased by 3.84 times compared to free tamoxifen citrate and 11.19 times compared to free tamoxifen when formulated in PLGA nanoparticles. Histopathological studies evidenced the low toxicity of tamoxifen encapsulated nanoparticles compared to free drug.[102]
ChitosanNPsAntigen (bovine serum albumin)Antigen-loaded chitosan nanoparticles; surface engineered with Ulex europaeus agglutinin lectin displayed efficient systemic and mucosal immune responses.[103]
PEG-poly(lactide) diblock copolymersMicelles Cyclosporine AEnhanced stability and intestinal absorption of cyclosporine A-loaded polymeric micelles were reported compared to commercial tablet formulation of cyclosporine A.[104]
ChitosanNPsRifampicinpH-dependent drug release (75%) at simulated intestinal pH over a period of 24 h[105]
Poly lactic acid-co-PEGMicellesFenofibrateMicelles having a size range of 158 to 249 nm were prepared as a carrier for oral administration.[106]
ChitosanMicellesPaclitaxel3.80-fold enhanced bioavailability of paclitaxel micelles compared to Taxol®[107]
Polyvinyl caprolactam-polyvinyl acetate-PEG graft copolymerMicellesSilybinSignificant increase in absorption of Silybin following oral administration of drug-loaded polymeric micelles in rats.[108]
Carbomer 934 and poloxamer P 407MicellesDeferoxamine mesylatePolymeric micelle exhibited 2.5 times increased drug permeation across intestine compared to control.[109]
N-naphthyl-N,O-succinyl chitosanMicellesCurcuminpH-responsive polymeric micelles exhibited a significantly increased amount of drug release in simulated colonic fluid compared to free drug.[110]
NPs, nanoparticles; NA, not applicable.
Table 3. A cross-section report on polymers employed as nanocarriers for ocular drug delivery.
Table 3. A cross-section report on polymers employed as nanocarriers for ocular drug delivery.
PolymerDelivery SystemTherapeutic MoleculeSummaryRef.
Eudragit® RL and RS (50:50)NPsGatifloxacinProlonged-release rate and antimicrobial activity. [117]
PLGA and PLGA–PEGNPsMelatoninPLGA–poly (ethylene glycol) loaded melanin displayed prolonged pharmacological effect (reduced intraocular pressure) up to 8 h.[118]
Chitosan-alginateNPsDaptomycinIn vitro ocular permeability study of daptomycin-loaded chitosan-alginate nanoparticles showed increased epithelial retention compared to free drug.[119]
PLGAwith Eudragit®RL or coated with Carbopol®NPsCyclosporin APolymeric nanoparticles displayed biphasic release i.e., initial burst followed by slow drug release up to 24 h. PLGA with Eudragit®RL showed the highest degree of cellular uptake, tear film concentration, and ocular bioavailability.[120]
PLGANPsPilocarpineChitosan-coated PLGA nanoparticles of pilocarpine displayed prolonged residence time after topical ocular application.[121]
PLGANPsMoxifloxacinPLGA nanoparticles loaded with moxifloxacin showed higher drug permeation compared to conventional eye drops. Sustained drug release was observed up to 24 h, thereby could avoid frequent administration of dosage.[122]
GelatinNPsMoxifloxacinMoxifloxacin-loaded gelatin nanoparticles exhibited burst release in the first hour followed by controlled release up to 12 h in an in vitro experiment.[115]
Sodium alginateNPsBrimonidine tartrateIn vivo experiment in albino rats displayed prolonged drug release up to 8 h following topical application of brimonidine tartrate loaded gelatin nanoparticles. [123]
ChitosanNPsBetaxolol hydrochlorideIn vitro drug release study showed an initial burst followed by sustained release up to 12 h. This could be due to mucoadhesiveness of chitosan leading to improved pre-corneal residence time and hence corneal permeability.[124]
Eudragit® RS 100NPsCyclosporin AIn vitro experiment of cyclosporin A loaded positively charged Eudragit® RS 100 nanoparticles displayed extended drug release. In vivo results showed prolonged residence time of drug and polymeric nanoparticles in vitreous humor.[125]
Poly-ε-caprolactone, 2-hydroxy ethyl methacrylate, PEG diacrylateNPsLoteprednolA drug release study of hydrogel-embedded polymeric nanoparticles showed extended release for up to 12 days.[126]
Chitosan-sodium alginateNPsGatifloxacinMucoadhesive polymeric nanoparticles showed fast release during 1 h followed by gradual release up to 24 h.[105]
Poly(beta-amino esters)NPsGenesPolymeric nanoparticles showed transfection efficiency of up to 85% for human reticuloendothelial cells and up to 65% for human umbilical vein endothelial cells.[127]
Poly(D,L-lactide co-glycolide) (PLGA)NPsLoteprednol etabonateEx vivo trans corneal permeation study across goat cornea revealed an improved permeation profile of formulated drug product compared to the plain dug owing to an increased residence time of PLGA nanoparticles.[128]
ChitosanNPsOrnidazoleIn vitro drug release study revealed initial burst release followed by gradual release up to 24 h of the ornidazole-loaded mucodhesive chitosan nanoparticles.[129]
ChitosanNPsNaringeninIn vitro study of naringenin-loaded chitosan nanoparticles revealed a moderate sustained-release effect. In vivo experiment exhibited prolonged residence time of polymeric nanoparticles compared to naringenin suspension which could be accountable for its improved bioavailability in aqueous humor.[130]
Methoxy poly(ethylene glycol)–poly(ɛ-caprolactone)MicellesNot applicableIn vitro studies of polymeric micelles did not show any cytotoxicity against human corneal epithelial cells, human lens epithelial cells, and retinal pigment epithelial cells at micellar concentrations of 0–2 mg/mL.[131]
Eudragit® RS100NPsVancomycinProlonged residence time and Cmax of vancomycin-loaded polymeric nanoparticle was observed resulting in a more than two-fold increment in bioavailability (AUC0.25–24) over control group.[132]
Poly(dl-lactide-co-glycolide) (PLGA)NPsSparfloxacinIn vitro release study exhibited an extended drug release profile. Gamma scintigraphy study in albino rabbits showed prolonged precorneal retention of the radiolabelled sparfloxacin-loaded polymeric nanoparticles compared to the marketed formulation. The formulation displayed non-irritant properties in the Hen egg test-chorioallantoic membrane test.[133]
Poly(ethylene oxide)-poly(propylene oxide)MicellesLornoxicamConfocal laser studies evidenced the appreciable corneal penetrating power of the polymeric micelles.[134]
Eudragit® RS100 and Eudragit® RL100NPsCloricromeneIn vitro studies showed modified release of drug from the polymer matrix.[135]
Poly(D,L-lactide-co-glycolide)NanospheresFlurbiprofenPolymeric nanospheres showed a two-fold increment in drug permeation compared to commercial eye drops formulation in an ex vivo experiment.[136]
Eudragit® RL100NPsAceclofenacIn vitro transcorneal permeability study across excised goat cornea revealed a 2-fold increment in drug permeation from polymeric nanoparticles compared to aqueous drug solution. No signs of corneal damage were reported.[137]
PLGA and chitosanNPsFluocinolone acetonidePolymeric nanoparticles showed good mucoadhesion characteristics and exhibited rapid and extended drug delivery to the eye evidenced in the pharmacokinetic experiment.[138]
Methylated β-cyclodextrinNPsFenofibratePolymeric NPs resulted in low cytotoxicity, low hemolytic potential, and moderately irritable to the eyes.[139]
Zein and hyaluronic acidNPsCiprofloxacinThe developed NPs were biocompatible, had high %EE, and prolonged release of the drug. These can be employed for the treatment of conjunctivitis.[140]
Chitosan NPsMeloxicamPolymeric nanoparticles showed sustained drug release behavior and improved permeation through the cornea.[141]
Poly(L-lactide)-b-poly(methacrylic acid-co-3-acrylamidophenylboronic acid)Polymeric micellesCyclosporin AThe developed micelles resulted in low cytotoxicity, reduction of dose, and improve bioavailability of the therapeutic molecule.[142]
SoluplusPolymeric micellesIbuprofen, idebenone, and miconazoleThe developed nanomicelles have potential applications in ocular delivery.[143]
PEGylated fatty acid-block-poly(ε-caprolactone)Polymeric micellesCyclosporin AThe developed nanomicelles showed sustained drug release behavior and can be employed for ocular drug delivery.[144]
PEG-b-PLAPolymeric micellesTriamcinolone acetonideThe developed micelles enhanced anti-inflammatory action. [145]
PLGAPolymeric nanospheresLactoferrinIn vitro and in vivo investigations revealed a significant increment in residence time of developed formulation on the eye surface.[146]
PLGA-PEGPolymeric nanospheresPioglitazoneThe ex vivo investigations of developed nanospheres revealed that permeation and retention via sclera were greater than corneal and non-irritant for the eye.[147]
NPs-Nanoparticles, PLGA—Poly(dl-lactide-co-glycolide), PEG—Poly(ethylene glycol), PLA—Polylactic acid.
Table 4. A summary of reports on polymers employed as nanocarriers for parenteral delivery of drugs, peptides, and genes.
Table 4. A summary of reports on polymers employed as nanocarriers for parenteral delivery of drugs, peptides, and genes.
PolymerDelivery SystemTherapeutic MoleculeSummaryRef.
PLGANPspDNA (alkaline phosphatase, a reporter gene)pDNA loaded in PLGA particles showed sustained release of pDNA while maintaining its structural and functional integrity for over a month.[101]
PCLNPsRisperidoneIn vivo studies evidenced the prolonged antipsychotic effect of risperidone-loaded biodegradable polymeric nanoparticles compared to risperidone solution administered through intravenous route.[151]
PLGANPsRisperidoneIn vivo studies in mice showed the prolonged antipsychotic effect of risperidone-loaded polymeric nanoparticles up to 72 h with fewer extrapyramidal side effects compared to risperidone solution administered subcutaneously.[152]
N-iso propylacrylamide, PVP, and acrylic acid in the ratio of 60:20:20NPsCurcuminPolymeric nanoparticles of curcumin displayed higher systemic bioavailability in plasma and tissues compared to free curcumin after parenteral administration. No systemic adverse action was reported.[153]
Sodium alginateNanoparticlesCarboplatinProlonged drug release up to 12 h compared to the pure drug (up to 3 h). The drug was detected in the liver, lungs, and spleen after parenteral administration in Laca mice, thereby showing the potential of sodium alginate nanoparticles as a promising tool for targeted drug delivery.[154]
Methoxypoly (ethylene glycol)-b-poly(ε-caprolactone-co-p-dioxanone)MicellesCurcuminPolymeric micelles showed slow drug release and dose-dependent inhibition of PC-3 human prostate cancer cells. [155]
PEG-b-PLA,
PEG-b-PCL
MicellesSagopiloneSagopilone-loaded polymeric micelles were found stable up to 24 h at 37 °C. In vitro studies showed high antiproliferative activity (IC50 < 1 nM). No carrier-related side effects were observed In vivo.[156]
PLGANanoparticlesZaleplonThe developed NPs showed greater ant-convulsant potential in contrast to free drug.[157]
Poly(propylenesulfide)NanoparticlesDiflunisalThe developed NPs resulted in a significant decline in S. aureus-mediated bone degradation and aided in establishing the possibility of systematic delivery of anti-viral agents to treat osteomyelitis. [158]
Chitosan and PCLNanoparticlesPaliperidoneThe developed formulations showed minimum cellular toxicity.[159]
Hexadecanol-modified chitosan oligosaccharideNanomicellesGambogic acidIt was reported that nanomicelles resulted in improved cellular uptake and quick drug release. [160]
Hyaluronic acid, vitamin E and styrene maleic anhydrideNanomicellesCurcumin analoguesThe developed nanomicelles revealed excellent TNBC accumulation with minimum spleen and liver retention.[161]
NPs, nanoparticles; PLGA, Poly(dl-lactide-co-glycolide); PEG, Poly(ethylene glycol); PLA, Polylactic acid.
Table 5. A cross-section of reports on polymers employed as nanocarriers for topical delivery of drugs, peptides, and genes.
Table 5. A cross-section of reports on polymers employed as nanocarriers for topical delivery of drugs, peptides, and genes.
PolymerDelivery SystemTherapeutic MoleculeSummaryRef.
ChitosanNPsRetinolEncapsulation of retinol in chitosan nanoparticles improved retinol stability and minimized its irritation and toxicity.[165]
PCLNPsIndomethacinEx vivo permeation study of polymeric nanoparticles displayed higher flux across human skin compared to marketed gel formulation, Indotopic® gel.[166]
PLGANPsAntisense oligodeoxynucleotidesPolymeric nanoparticles displayed 25 to 32% drug release within one day via a diffusion-controlled process followed by PLGA degradation-controlled drug release (39% to 70%) after 14 days.[167]
GelatinNPsSTAT6 siRNAGelatin nanoparticles exhibited stable and biocompatible formulation for topical delivery of siRNA.[168]
Ethyl cellulose, Eudragit® RSNanospheres/NanocapsulesDexamethasoneIn an ex vivo study, polymeric nanoparticles displayed slower drug release and penetration compared to conventional cream and could be an efficient way to control the release and penetration of dexamethasone on the skin and mucous membrane.[169]
HPMC K15 & PEG 200NPsRosuvastatinThe ex vivo permeation study exhibited significantly higher permeation via rat skin.[170]
Table 6. USFDA-approved polymeric products.
Table 6. USFDA-approved polymeric products.
NameFormulationIndicationsRoleRef
Polymeric nanoparticles
Cimzia®PEGylated antibody fragment (Certolizumab)Crohn’s disease, rheumatoid arthritis, Ankylosing spondylitisImproved circulation time and greater stability In vivo.[177]
Krystexxa®Polymer-protein conjugate (PEGylated porcine-like uricase)Chronic goutImproved stability of protein through PEGylation[178]
Plegridy®Polymer-protein conjugate (PEGylated INF)Multiple sclerosisImproved protein stability by PEGylation[179]
AdenovatePolymer-protein conjugate (PEGylated Factor VIII)Hemophilia[180]
Neulasta®PEGylated GCSF proteinNeutropenia[181]
Pegasys®PEGylated IFN alpha-2a proteinHepatitis B and C[182]
Copaxone®Copolymer of L-glutamic acid, L-alanine, L-lysine, and L-tyrosineImmunomodulator in multiple sclerosisImproved biocompatibility/solubility[183]
Polymeric micelles
Estrasorb™Micellar estradiolMenopausal therapyControlled delivery[184]
Table 7. A cross-section of reports on polymeric nanocarriers under clinical trials.
Table 7. A cross-section of reports on polymeric nanocarriers under clinical trials.
Product DevelopedTherapeutic AgentTargeted DiseaseObjectiveClinical Trial Status/Verification Date
Polymeric nanoparticle surface modified with somatostatin analogCetuximabColon cancer and colorectal cancerTo evaluate the bioavailability and therapeutic window.Phase 1 (Recruiting)/October 2019
Polymeric micelleDocetaxelEsophageal CarcinomaTo determine the effects and safety. Phase 2 (Recruiting)/July 2018
Polymeric micellePaclitaxelRecurrent breast cancerTo examine the response rate.Phase 4/June 2009
Polymer basednanoparticlesDocetaxelAdvanced solid malignanciesTo determine the maximum tolerated dose and evaluate the safety and pharmacokineticsPhase 1 (completed)/May 2017
Polymeric micelleDocetaxelHead and neck squamous cell carcinomaTo determine safety and efficacyPhase 2 (Recruiting)/April 2017
Polymeric micellePaclitaxelOvarian cancerTo determine the maximum tolerated dose and evaluate the safety/efficacyPhase 1 & 2/Dec 2009
Polymeric micellePaclitaxelNon-small cell lung cancerTo examine the response rate.Phase 2 (completed)/May 2017
Polymeric micellePaclitaxelBladder cancer and ureter cancerTo examine safety and efficacyPhase 2 (completed)/December 2011
Table 8. A random sample of patent reports on polymeric nanocarriers intended for oral, ocular, topical, and parenteral administration.
Table 8. A random sample of patent reports on polymeric nanocarriers intended for oral, ocular, topical, and parenteral administration.
Patent Number,
Year
Title of the PatentDescription
US 20150353676 A1,
2015
Polymeric nanoparticles and a process of preparation thereofDisclosed composition and method of preparation for emulsifier-free biodegradable polymeric nanoparticles made of a block copolymer having a size range between 30–120 nm.
US 20150320856 A1,
2015
Method for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance immune responsesDescribed composition and method of preparation for immunosuppressant loaded pH-sensitive polymeric synthetic nanocarrier, surface engineered with APC antigen for site-specific and controlled drug delivery.
US 20160324966 A1,
2016
Polymeric nanocarriers with a light-triggered release mechanismHighlighted a method for light-triggered release of PLGA polymeric nanocarrier for biomedical applications.
US 20150079005 A1,
2015
Polymeric nanocarriers with a linear dual response mechanismDepicted a method for preparation of pH-sensitive polymeric nanocarrier which degrades at lower pH and/or reactive oxygen species.
US 20090258078 A1,
2009
Antioxidant polymer nanocarriers for use in preventing oxidative injuryPresented a composition and preparation method for protein encapsulated polymeric nanocarrier with an intent to protect the protein from protease degradation and increase the therapeutic half-life for topical delivery.
US8613951 B2,
2013
Therapeutic polymeric
nanoparticles with mTOR inhibitors and methods of making and using the same
Reveals a preparation method for mTOR inhibitors loaded polymeric nanoparticles made of the diblock copolymer.
US8715741 B2,
2014
Water-dispersible oral parenteral, and topical formulations for poorly water-soluble drugs using smart polymeric nanoparticlesDiscloses composition for poorly water-soluble drug encapsulated polymeric nanocarrier conversant with mucoadhesive, oral bioavailability and multifunctional systemic targeting characteristics.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tewari, A.K.; Upadhyay, S.C.; Kumar, M.; Pathak, K.; Kaushik, D.; Verma, R.; Bhatt, S.; Massoud, E.E.S.; Rahman, M.H.; Cavalu, S. Insights on Development Aspects of Polymeric Nanocarriers: The Translation from Bench to Clinic. Polymers 2022, 14, 3545. https://doi.org/10.3390/polym14173545

AMA Style

Tewari AK, Upadhyay SC, Kumar M, Pathak K, Kaushik D, Verma R, Bhatt S, Massoud EES, Rahman MH, Cavalu S. Insights on Development Aspects of Polymeric Nanocarriers: The Translation from Bench to Clinic. Polymers. 2022; 14(17):3545. https://doi.org/10.3390/polym14173545

Chicago/Turabian Style

Tewari, Akhilesh Kumar, Satish Chandra Upadhyay, Manish Kumar, Kamla Pathak, Deepak Kaushik, Ravinder Verma, Shailendra Bhatt, Ehab El Sayed Massoud, Md. Habibur Rahman, and Simona Cavalu. 2022. "Insights on Development Aspects of Polymeric Nanocarriers: The Translation from Bench to Clinic" Polymers 14, no. 17: 3545. https://doi.org/10.3390/polym14173545

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