Next Article in Journal
Fluro-Protein C-Phycocyanin Docked Silver Nanocomposite Accelerates Cell Migration through NFĸB Signaling Pathway
Next Article in Special Issue
ZnO Tetrapods for Label-Free Optical Biosensing: Physicochemical Characterization and Functionalization Strategies
Previous Article in Journal
Cuban Sugar Cane Wax Alcohol Exhibited Enhanced Antioxidant, Anti-Glycation and Anti-Inflammatory Activity in Reconstituted High-Density Lipoprotein (rHDL) with Improved Structural and Functional Correlations: Comparison of Various Policosanols
Previous Article in Special Issue
Employing Supervised Algorithms for the Prediction of Nanomaterial’s Antioxidant Efficiency
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sustainable Biodegradable Biopolymer-Based Nanoparticles for Healthcare Applications

1
Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova Ulica 17, 2000 Maribor, Slovenia
2
Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(4), 3188; https://doi.org/10.3390/ijms24043188
Submission received: 29 December 2022 / Revised: 1 February 2023 / Accepted: 2 February 2023 / Published: 6 February 2023
(This article belongs to the Special Issue Functional Nanomaterials for Healthcare)

Abstract

:
Biopolymeric nanoparticles are gaining importance as nanocarriers for various biomedical applications, enabling long-term and controlled release at the target site. Since they are promising delivery systems for various therapeutic agents and offer advantageous properties such as biodegradability, biocompatibility, non-toxicity, and stability compared to various toxic metal nanoparticles, we decided to provide an overview on this topic. Therefore, the review focuses on the use of biopolymeric nanoparticles of animal, plant, algal, fungal, and bacterial origin as a sustainable material for potential use as drug delivery systems. A particular focus is on the encapsulation of many different therapeutic agents categorized as bioactive compounds, drugs, antibiotics, and other antimicrobial agents, extracts, and essential oils into protein- and polysaccharide-based nanocarriers. These show promising benefits for human health, especially for successful antimicrobial and anticancer activity. The review article, divided into protein-based and polysaccharide-based biopolymeric nanoparticles and further according to the origin of the biopolymer, enables the reader to select the appropriate biopolymeric nanoparticles more easily for the incorporation of the desired component. The latest research results from the last five years in the field of the successful production of biopolymeric nanoparticles loaded with various therapeutic agents for healthcare applications are included in this review.

1. Introduction

The field of nanotechnology, which encompasses the synthesis, development, and application of nanomaterials in a size range from 1 to 100 nm, has gained increasing interest in various areas in recent years. Among other things, nanomaterials, especially nanoparticles, play an important role, especially in the field of healthcare applications, where they offer higher efficiency and lower toxicity [1,2,3].
Treatment of various life-threatening diseases, such as chronic diseases (e.g., cancer, heart disease, HIV/AIDS, diabetes) and severe neurological and infectious diseases, is becoming increasingly challenging. Nanomaterials are successfully addressing these challenges by finding new ways for potential treatment, intending to overcome the obstacles of conventional treatment forms [1,2,4]. Various existing formulations for the treatment of certain diseases are poorly soluble, resulting in minimal systemic bioavailability, unstable in vivo, reducing the efficacy of the drug, and causing toxic side effects, even to normal cells. Furthermore, they can cause nephrotoxicity, neurovirulence, and gastrointestinal reactions. Drug resistance may also occur, reducing the impact of further treatment [5].
Nanomaterials are defined as materials with at least one dimension smaller than 100 nm [2,6]. Compared to bulk materials, nanomaterials have different physicochemical properties, such as different shapes, surface area, size, and reactivity, leading to their exceptional performance in many applications. Nanomaterials can be classified according to different criteria [7,8]. One is the classification into four different categories depending on the dimensionality, as shown in Figure 1. Zero-dimensional nanomaterials (0–D) include nanoparticles, fullerenes, and quantum dots. All their three dimensions are in the nanometer range. In one-dimensional nanomaterials (1–D), one dimension is outside the nanoscale, while the other two are inside the nanoscale. This classification includes nanofibers, nanorods, nanotubes, nanowires, and nanohorns. Two-dimensional nanomaterials (2–D), such as nanofilms, nanosheets, and nanolayers, have two dimensions beyond the nanoscale. The most common examples of three-dimensional nanomaterials (3–D) or bulk nanomaterials are core shells, arrays of nanotubes, and nanowires. In this category, the materials are not in the nanoscale in any dimension [2,6,9,10,11,12].
Nanotechnology-based formulations enable better, more effective, and safer use of drugs for the prevention and treatment of various conditions, which is why the production of new nanocarriers for therapeutic agents is becoming increasingly important [1].
A variety of nanoparticles with unique physical and chemical characteristics hold promise for disease diagnosis, biological imaging, and nanomedicine applications [1,14]. They provide the ability to protect the drug from premature degradation, improve intracellular penetration, prolong release, increase absorption and efficacy of the drug at the target site, and therefore represent significant potential in the healthcare sector [4].
It is important to note that the most crucial factor in the production of nanoparticles is to avoid biological and physical barriers such as diffusion, protein adsorption, aggregation, renal or hepatic clearance, and phagocytic sequestration [14].
An important advantage of nanoparticles is that they can be reused after the removal of the incorporated component, which is an important contribution to the circular economy from an economic point of view [15].
Nanoparticles can also be divided according to their composition, meaning according to the material from which they are produced. These include in particular organic, inorganic, and carbon-based nanoparticles (Figure 2) [16]. Carbon-based nanoparticles consist only of carbon atoms and have different morphologies. They are divided into fullerenes, graphene, carbon black, carbon quantum dots, carbon nanotubes, and nanodiamonds [2,7,17,18]. Non-carbon nanoparticles are inorganic nanoparticles and can be produced from pure metals (Au, Ag, Al, Zn, Fe, Cd), metal oxides (ZnO, TiO2, MnO2, Fe2O3, Al2O3), semiconductors (CdS, ZnS), and ceramics (SiO2, ZrO2) [7,16,19]. Organic nanoparticles include proteins, lipids, polymers, carbohydrates, or other organic compounds and can be classified as synthetic or natural. They are biocompatible, biodegradable, generally non-toxic, and improve pharmacokinetic properties, bioavailability, and drug targeting. Examples are liposomes, polymersomes, micelles, polymer constructs, and protein complexes [2,19,20].
Inorganic nanoparticles, especially metal- and metal oxide-based nanoparticles, are among the most popular. In various applications, such as drug delivery, they are characterized by good stability and half-life of the carrier in the bloodstream, adequate biodistribution, and targeted delivery of the drug to the desired target site [21]. For example, gold nanoparticles are used for numerous diagnostic purposes [22], and silver nanoparticles are also important in biomedicine, primarily because of their antimicrobial potential [23]. Among the metal oxide nanoparticles, ZnO nanoparticles should be highlighted, which are biocompatible, non-toxic, non-expensive, and provide effective antibacterial activity and UV protection [24].
However, they are harmful to the environment, soil, water, and the organisms in them. In addition, some can be very toxic to the human body. Therefore, their use is limited because the body cannot metabolize and excrete them [3]. In contrast, biopolymeric nanoparticles are suitable for various healthcare applications due to their biocompatibility, biodegradability, nontoxicity, and other advantageous properties. The review article covers the latest research results of the last five years on nanoparticles based on natural biopolymers such as proteins and polysaccharides of animal, plant, algal, fungal, and bacterial origin. Special emphasis is paid to the incorporation of various therapeutic agents such as bioactive agents, conventional drugs, antibiotics, and other antimicrobial agents, extracts, and essential oils into biopolymeric nanoparticles. These thus represent promising delivery systems that prevent potential adverse side effects of therapeutic agents, enable targeted delivery to target sites, and thus contribute to enhanced therapeutic efficacy and sustained release.

2. Polymeric Nanoparticles

Drug delivery systems must meet specific criteria, such as being biocompatible, biodegradable, stable, non-toxic, non-immunogenic, cost-effective, selective, easy to produce, and delivering and releasing drugs only at specific locations. When using free dosage forms, not only diseased but also healthy cells, tissues, organs, or subcellular organs can be damaged during drug administration and release. Various delivery systems, such as nanoparticles with encapsulated drugs, can successfully circumvent these limitations (Figure 3) [25,26,27]. Drug delivery systems also make it possible to achieve the desired therapeutic effect of drugs and, at the same time, greater efficiency and safety, as well as controlled release of the drug at the target site and prevent aggregation and reduce possible side effects of delivered drugs [25,28,29].
Polymer-based nanoparticles are important as alternative carriers for various therapeutic agents for different medicinal purposes mainly due to their biodegradability, biocompatibility, non-immunogenicity, and non-toxicity [29]. Drugs can be incorporated into polymeric nanoparticles by physical or chemical means, i.e., by various adsorption processes on their surface after the nanoparticle formation or by incorporation of drugs into the nanoparticles during the production process. The loading efficacy of therapeutic agents on or into nanoparticles is mainly influenced by nanoparticle size, molecular weight and solubility of drugs, and chemical interactions between drugs and nanoparticles. The release rate of therapeutic agents from polymeric nanoparticles is primarily based on the desorption of surface-bound or adsorbed therapeutic agents, diffusion of therapeutic agents from polymeric nanoparticles, erosion of polymeric nanoparticles, and the combined process of diffusion and erosion [31,32]. Polymeric nanoparticles offer several advantages. They increase the absorption of loaded drugs, provide protection against degradation, prolong the circulation time, and ensure delivery to the target site due to their good solubility, safety and stability, and long-term release [33]. They can be of synthetic or natural origin. However, synthetic polymers can be toxic and immunogenic. Therefore, natural and biodegradable polymers offer better opportunities for nanoparticle production for various biomedical applications [29,34]. Figure 4 shows the advantages and disadvantages schematically and provides examples of synthetic and natural polymers.

2.1. Biopolymers Used for the Preparation of Biodegradable Nanoparticles

The most favorable nanoparticles for medical use must meet certain requirements, such as improved bioavailability, protection of therapeutic agents from degradation, controlled drug release, specific targeting, minimal toxicity, stability, and low cost [38], and biopolymeric nanoparticles best meet these requirements.
Biopolymeric nanoparticles comprise biopolymers—natural biological materials that are also generally recognized as safe (GRAS). Biopolymers are macromolecules composed of repeating units of monomers linked together by covalent bonds. Biopolymers such as proteins and polysaccharides are suitable for the preparation of biodegradable polymeric nanoparticles for various biomedical applications, especially for drug delivery, tissue engineering, and wound healing, as the therapeutic effect of the incorporated bioactive substances can be enhanced, and potential side effects can be reduced. This is due to their unique functions and properties, including biodegradability, biocompatibility, photoprotection, and antibacterial and antioxidant properties. In addition, they are naturally renewable, relatively inexpensive, and provide high stability in biological fluids and during storage [3,39,40,41,42,43]. Furthermore, various drugs and therapeutic agents can be successfully incorporated into biopolymeric nanoparticles, contributing to enhanced release at the target site [44]. Both natural polysaccharides and proteins are appropriate carriers for various therapeutic agents in the biomedical field [45]. The use of polysaccharide–protein complexes and conjugates (protein–protein, polysaccharide–polysaccharide, and protein–polysaccharide composite nanoparticles) may be even more efficient due to their additional enhanced properties. Biopolymer composite nanoparticles offer many opportunities to improve biological functions. The preparation of protein–polysaccharide complexes can be manipulated to exhibit improved biocompatibility, biodegradability, stability, targeting, and mechanical properties [46].
Biopolymers can be obtained from various natural sources [37]. Depending on their origin, they can be divided into the animal, plant, algal, fungal, and bacterial biopolymers. Figure 5 shows a schematic representation of the biopolymers classification that can be used for the preparation of biopolymeric nanoparticles, depending on their origin, including the main representatives.
Due to their biodegradability, biocompatibility, nontoxicity, and stability, biopolymeric nanoparticles are promising nanocarrier systems for many drugs and other therapeutic agents. Through different production methods, biopolymeric nanoparticles can be successfully produced. The most commonly used methods are shown schematically in Figure 6. However, the choice of method plays a crucial role in the synthesis of nanoparticles with suitable properties, as it depends on the biopolymer used, the type of therapeutic agent, and the possible route of administration [44,49,50].
The desolvation method is based on using a desolvating (dehydrating) agent, such as ethanol or acetone, which changes the structure of the polymer and reduces its solubility, resulting in the formation of a precipitate of polymeric particles. This method is most commonly used to prepare protein-based nanoparticles [51,52,53]. The self-assembly method depends on the formation of biopolymeric nano-systems by self-assembly, both naturally and artificially, through non-covalent bonds [54,55]. The emulsification method is based on the preparation of nanoparticles by forming an emulsion system by mixing two immiscible liquids, namely the organic and aqueous phases, at very high shear. The organic solvent is then removed by evaporation to form solid nanoparticles [54,56,57]. The nano-spray drying method transforms the material from a liquid into a powder. The solution is sprayed into small droplets through a nozzle into a chamber where drying is carried out by hot gas. The evaporation of the solvent forms dry particles that collect at the bottom of the chamber. The electrospray method is based on manipulating materials on the submicron scale. The polymer solution is sprayed using high voltage through a capillary nozzle that forms aerosolized droplets containing polymer nanoparticles of colloidal size [52,54,58,59]. The complex coacervation method uses pH-dependent electrostatic interactions between biopolymers to form stable nanoparticles and coacervates [52,60]. The ionic gelation method depends on electrostatic interactions between charged polysaccharides in an aqueous medium under certain conditions. It is considered the most commonly used method for the preparation of stable polysaccharide nanoparticles [3,61,62,63].

2.1.1. Protein Nanoparticles

Proteins are biological macromolecules with a high molecular weight, which are inexpensive, abundant, and renewable. Their unique functionalities and properties make them ideal natural materials for the preparation of biodegradable polymeric nanoparticles. They are amphiphilic, which allows good interactions with solvents and various drugs. Protein nanoparticles are produced under mild conditions that do not require the use of toxic chemicals or organic solvents [54,64]. They offer many advantages because they are biocompatible, biodegradable, stable, have non-antigenic properties, are non-immunogenic and non-toxic, and safe.
Moreover, surface modifications of nanoparticles are also possible [52,65]. They are suitable as carriers for various substances, such as natural or chemical therapeutics, growth factors, peptides, vaccines, and nucleic acids. In this way, the potential side effects of the drugs are eliminated, while at the same time, their therapeutic effect at the target site is increased [56]. Therefore, they can be successfully used in various biomedical applications, especially for tissue engineering and the delivery of drugs and other therapeutics [65,66].
Furthermore, various innovative protein-based nanoparticles offer enhanced properties such as biocompatibility, stability, etc. For example, intrinsically disordered proteins (IDPs) are promising nanomaterials for various medical and biotechnological purposes. They can help improve the development of drug delivery systems by enhancing the self-assembly, stimuli-responsiveness, and recognition properties of protein/peptide copolymers [67,68,69,70].
Protein-based nanoparticles can be classified according to the origin of the proteins used for their production. Proteins can be isolated from various animal and plant species. The most commonly used proteins are, for example, collagen, gelatin, and keratin. The composition of proteins depends mainly on the species from which they are obtained. Table 1 lists the most important examples of proteins and their origins suitable for protein-based nanoparticles production.

2.1.2. Polysaccharide Nanoparticles

Polysaccharides are natural biopolymeric materials suitable for biopolymeric nanoparticle fabrication due to their exceptional structural characteristics. They consist of monosaccharide units linked together by glycosidic bonds [79]. Polysaccharides have many advantageous properties, such as biocompatibility, biodegradability, and nontoxicity. In addition, they are relatively inexpensive and readily available [79,80,81]. The presence of hydroxyl groups and other hydrophilic groups, such as carboxyl (e.g., in alginate) and amino groups (e.g., in chitosan), allows easy modification and functionalization of many types of polysaccharides for specific purposes [82,83].
Due to various favorable properties, polysaccharide-based nanoparticles are suitable for biomedical applications such as drug delivery and tissue engineering. In addition, they are successfully used as delivery vehicles for different therapeutic agents, including proteins, peptides, and nucleic acids. They can ensure high encapsulation efficiency of various substances and rapid and controlled release at the target site. Furthermore, polysaccharide-based biomaterials have the potential to be used in skin tissue engineering because they can absorb a large amount of water. Due to their biocompatibility and good mechanical properties, they are also increasingly used for bone and cartilage tissue engineering and are also important for the tissue engineering of nerves [80,84].
Polysaccharide nanoparticles can be divided according to the origin of the polysaccharides used for their production [85]. Polysaccharides can be isolated from animal, plant, algal, fungal, and bacterial species. They have different biological characteristics and activities due to different chemical structures and ionic nature [61]. The most well-known natural and biodegradable polysaccharides used in healthcare applications are chitosan, alginate, carrageenan, gellan gum, and dextran [86,87].
Table 2 lists the most important examples of polysaccharides and their origins used for polysaccharide-based nanoparticles production.

3. Animal-Based Biopolymeric Nanoparticles Used for Healthcare Applications

Biopolymers of animal origin, such as proteins and polysaccharides, play an essential and versatile role in various biomedical applications, including the production of nanoparticles, especially for drug delivery and tissue engineering.

3.1. Protein Nanoparticles of Animal Origin

For the use of biopolymeric nanoparticles for various biomedical applications as carriers of numerous therapeutic substances, proteins of animal origin are most commonly used, of which albumin, casein, collagen, gelatin, keratin, and silk proteins (fibroin and sericin) are the best known.

3.1.1. Albumin

Albumin is the main protein in blood plasma, and due to its many favorable properties, it is a versatile and suitable biomaterial for the preparation of biopolymeric nanoparticles. It is biodegradable, biocompatible, non-toxic, non-immunogenic, available in pure form, highly soluble in water and physiological fluids, and stable in the pH range between 4.0 and 9.0. It also exhibits thermal stability when heated to 60 °C for up to 10 h, as it does not cause denaturation. Albumin can be obtained from a variety of sources, including human, bovine, rat, and chicken, with bovine serum albumin (BSA), human serum albumin (HSA), egg white albumin (ovalbumin), etc. Albumin-based nanoparticles can successfully contribute to increased bioavailability, improved pharmacokinetic properties, and enhanced ability to release incorporated drugs at target sites [51,56,71,128,129].
They are successfully used as nanocarriers in the treatment and diagnosis of various diseases, as they can effectively and safely deliver nonspecifically distributed drugs in the body which can cause serious side effects [71,130]. Albumin-based nanoparticles have already been developed for the treatment of cancer, diabetes, hepatitis C, and arthritis [131,132]. Yan et al. [130] prepared albumin nanoparticles loaded with prednisolone and curcumin using a high-pressure homogenization method. Combining two therapeutic substances loaded into albumin-based biopolymeric nanoparticles has shown promise as a delivery system for combined chemotherapy in rheumatoid arthritis. By using biopolymeric nanoparticles, a better anti-inflammatory effect against macrophages was obtained than with a free mixture of drugs. Gawde et al. [133] came to similar conclusions that albumin nanoparticles with incorporated paclitaxel and difluorinated curcumin are suitable for the treatment of gynecologic cancers. Zhao et al. [134] also demonstrated that albumin-based nanoparticles are potential carriers for the co-delivery of paclitaxel and resveratrol for effective synergistic cancer treatment. Moreover, albumin-based nanoparticles produced by the desolvation method with incorporated methylene blue can effectively inhibit the growth of the yeast Candida albicans and reduce biofilm formation, as demonstrated in a study by Ambrósio et al. [135]. Using the desolvation method, the most commonly used technique for the preparation of albumin-based nanoparticles, nanoparticles with uniform size, spherical shape, and low aggregation tendency can be produced [135].

3.1.2. Collagen

Collagen is a natural biopolymer and the most abundant protein in the human body, namely the primary material of the extracellular matrix of bones, skin, muscles, cartilage, ligaments, and tendons. It accounts for more than 30% of all body proteins. Due to numerous advantages such as biocompatibility, biodegradability, non-immunogenicity, non-toxicity, tensile strength, low antigenicity, extensibility, and accessibility, it plays an essential role in the synthesis of nanoparticles in various biomedical fields, especially in drug delivery and tissue engineering [136,137,138]. Biodegradable and thermally stable collagen nanoparticles are suitable nanocarriers for many therapeutic and cytotoxic substances, genes, proteins, growth factors, and drugs. In dermatology, for example, they are important for administering antimicrobials and steroids. They have good absorption capacity, large contact surface area, and ensure controlled and long-term release of drugs [136,139,140].
Vijayakumar and Vaseeharan [141] synthesized non-toxic and environmentally friendly collagen-based ZnO nanoparticles with antimicrobial and antibiofilm activity against Gram-positive bacteria Streptococcus mutans and Gram-negative bacteria Proteus vulgaris and yeast C. albicans, and good anticancer activity against human hepatoma cancer cells (HepG2) at a very low concentration (75 μg/mL). Due to their biocompatibility and potential to mimic the extracellular matrix, collagen nanoparticles play an essential role in tissue engineering and regenerative medicine, especially in bone, cartilage, dental, vascular, corneal, and skin tissue engineering, wound healing, spinal cord injury repair, and nerve and oral mucosa regeneration [137,142,143]. In a study by Mondal et al. [144], the chemotherapeutic drug doxorubicin was successfully incorporated into gold-loaded collagen hydroxyapatite nanoparticles. The synthesized nanoparticle system is a promising material for drug delivery and an extracellular matrix for tissue engineering.

3.1.3. Other Animal-Based Protein Nanoparticles

One other important biopolymer for producing nanoparticles of animal origin, which is also promising for nanoparticle fabrication to improve bioavailability, efficacy, and controlled release, is casein, the main protein in milk and a good source of amino acids. Various therapeutic substances, such as the anticancer drug doxorubicin [145], the polyphenolic compounds resveratrol [146], curcumin [147] and quercetin [148], the antibacterial agent mequindox [149], and the phenylpropanoid eugenol [150], have already been successfully incorporated into casein nanoparticles for use in various healthcare applications. Gelatin is another natural, water-soluble protein. Various anticancer drugs, anti-inflammatory drugs, analgesics, antimicrobial agents, and other drugs have already been successfully loaded into gelatin nanoparticles [139,151]. Keratin is a protein rich in amino acids and is fibrous and insoluble. Keratin-based nanoparticles are suitable carriers for various antitumor drugs such as doxorubicin, paclitaxel, and chlorine c6 and 9(R)-9-hydroxystearic acid, and multiple polyphenols such as rutin, quercetin, and curcumin. In addition, they are also crucial for tissue engineering and wound healing [152,153]. Fibroin and sericin are the major proteins of silk proteins. Silk protein nanoparticles are also favorable delivery systems for many drugs, such as paclitaxel, 5-fluoroacil, doxorubicin, gemcitabine, fenofibrate, atorvastatin, methotrexate, inulin, resveratrol, and curcumin [154,155].

3.2. Polysaccharide Nanoparticles of Animal Origin

Polysaccharides of animal origin can be successfully used for various healthcare applications, with chitosan being the most important and well-known representative. It is a promising natural biomaterial for the synthesis of nanoformulations for the delivery of different therapeutic agents.

Chitosan

Chitosan is a natural biopolymer composed of N-acetyl-D-glucosamine and D-glucosamine units linked by β-1,4-glycosidic bonds. It is a cationic polysaccharide derived from chitin by demineralization and deproteinization. This is found mainly in the exoskeleton of crustaceans and insects, fish scales, and in the cell wall of fungi. Due to its exceptional physicochemical properties such as biodegradability, biocompatibility, non-toxicity, low immunogenicity, mucoadhesiveness, low cost, bio-renewability, and environmental friendliness, chitosan plays an important role in various biomedical and pharmaceutical applications. In addition, they have antitumor, antioxidant, and antimicrobial properties against the growth of Gram-positive and Gram-negative bacteria. Chitosan nanoparticles are stable, increase bioavailability, improve the solubility of poorly soluble drugs, and enhance the targeted delivery of various therapeutic compounds. Chemical modification or integration with functional materials has positive properties to improve in vivo release stability [156,157,158,159,160,161,162,163]. Chitosan-based nanoparticles can be used as nanocarriers for various drug antibiotics, proteins, vaccines, and genes [164,165]. They can be administered by nasal, intravenous, oral, and ocular routes [166].
For example, Ansari et al. [167] synthesized chitosan-based nanoparticles loaded with zinc gluconate using an ionic gelation method. They possess significant antioxidant, anti-inflammatory, and anti-arthritic potential, making them a potential alternative for the treatment of rheumatoid arthritis. Tzeyung et al. [168] prepared rotigotine-loaded chitosan nanoparticles for nose-to-brain delivery using an ionic gelation method. The nanoformulation did not cause toxicity or structural damage to the nasal mucosa, making chitosan-based nanoparticles an effective drug carrier for nose-to-brain delivery as an alternative to conventional administration routes. Yu et al. [169] synthesized dexamethasone–glycol chitosan nanoparticles that exhibited good ocular tolerance and had relatively longer precorneal retention than an aqueous solution. Therefore, these nanoparticles are relevant for ophthalmic drug administration in the treatment of various inflammatory diseases. Chitosan nanoparticles and chitosan/collagen peptide nanoparticles have been shown to be effective biopolymeric stabilizers of Pickering emulsions used for cosmetic applications [170,171]. In addition, chitosan nanoparticles have already been shown to be excellent inhibitors of bacteria, both Escherichia coli and Staphylococcus aureus [172]. Recent findings include the use of chitosan in the preparation of nanocarriers to enhance the antimetastatic properties of the entrapped drug in vitro and in vivo [173].
Table 3 summarizes examples of biopolymeric nanoparticles of animal origin for various healthcare applications, including their preparation method and the size of the synthesized nanoparticles.

4. Plant-Based Biopolymeric Nanoparticles Used for Healthcare Applications

Plants are a rich source of many vital substances, including various polysaccharides and proteins, promising biomaterials for producing biopolymeric nanoparticles for various biomedical applications.

4.1. Protein Nanoparticles of Plant Origin

Protein nanoparticles of plant origin are of great importance in the production of biopolymeric nanoparticles because they offer a significant advantage over protein nanoparticles of animal origin. Not only are they much cheaper and widely available, but they are also safer, as diseases from animals can be transmitted to humans through nanoparticles of animal origin. The most important representatives include zein and gliadin.

4.1.1. Gliadin

Gliadin is an important natural protein found in wheat gluten. It can be divided into four classes, namely α-, β-, γ-, and ω-gliadin, according to electrophoretic mobility. The structure of gliadin consists of a terminal region, namely a short N-terminal domain and a C-terminal domain, which is more hydrophobic due to the presence of amino acids, and a central repetitive region rich in glutamine and proline, which confers amphiphilicity to gliadin. Due to its relevant properties, it is suitable for the synthesis of nanoparticles for various therapeutic applications. They are biocompatible, biodegradable, non-toxic, and stable. Due to their hydrophobic nature and low solubility in aqueous solutions, they provide protection and controlled release of the loaded drugs [56,185,186,187]. Gliadin nanoparticles are suitable nanocarriers for various hydrophilic therapeutic agents [188]. Hydrophilic and amphiphilic compounds such as vitamins A and E and amoxicillin have already been successfully incorporated into gliadin nanoparticles [139].
In the study, Alqahtani et al. [189] confirmed that gliadin nanoparticles prepared by a modified antisolvent nanoprecipitation method were compatible with blood regardless of the size of the synthesized nanoparticles. Wu et al. [190] prepared potential delivery agents, resveratrol-incorporated gliadin nanoparticles, by an antisolvent precipitation method and stabilization with gum Arabic and chitosan hydrochloride. The nanocarriers prepared in this way significantly increased the release of resveratrol in water and the simulated digestive tract, thereby increasing its bioavailability. Moreover, they improved the chemical stability and antioxidant activity of the encapsulated polyphenolic compound. Sharif et al. [191] prepared zein nanoparticles with incorporated γ-oryzanol by electrospray method. Due to the encapsulation of γ-oryzanol, the synthesized nanoparticles were rounder and smoother with a homogeneous distribution of γ-oryzanol. The synthesized γ-oryzanol-loaded gliadin nanoparticles had improved stability and decreased the proliferation of HT-29 human colon cancer cells.

4.1.2. Zein

Zein is a major plant-based protein obtained mainly from corn. It is rich in prolamin, poorly soluble in water, and resistant to high temperatures. It is divided into four classes according to different peptide chains, solubility, and molecular size, namely α-, β-, γ-, and δ- zein. Zein is biodegradable, biocompatible, and safe and therefore promising for the synthesis of natural biopolymeric nanoparticles for various healthcare applications. Namely, it offers greater bioavailability, protection, and prolonged, controlled, and more effective release of incorporated substances at the target site. Many therapeutic agents have been successfully loaded into zein-based nanoparticles, e.g., doxorubicin, heparin, 5-fluorouracil, coumarin, quercetin, resveratrol, vitamin D3, and different antimicrobials [185,192,193,194].
Zein-based nanoparticles are applicable nanocarriers for hydrophobic molecules to enhance their bioavailability for various therapeutic purposes. Nunes et al. [195] successfully prepared zein nanoparticles using the nanoprecipitation technique, into which they incorporated resveratrol with an efficiency of 77%. The resveratrol-loaded zein nanoparticles successfully protected resveratrol from enzymatic degradation and showed low toxicity to human colorectal cell lines Caco-2 and HT29-MTX. Using a modified phase separation method, Zhang et al. [193] prepared dodecamer peptide (G23)-functionalized polydopamine (pD)-coated curcumin-loaded zein nanoparticles. The prepared spherical nanoparticles with a diameter of about 120 nm showed excellent cellular uptake by C6 glioma cells and the ability to penetrate 3D tumor spheroids. Zein is also widely used in tissue engineering [192]. Hadavi et al. [196] synthesized zein nanoparticles as a delivery system for a bone morphogenic protein-6 (BMP6) derived peptide using a liquid–liquid phase separation method. They are promising in bone regeneration due to their low toxicity, high encapsulation efficiency, and osteoinductivity.

4.2. Polysaccharide Nanoparticles of Plant Origin

Polysaccharides are found in large amounts in various plant species and, due to their non-toxicity compared to various synthetic materials, represent an important natural and biodegradable material for the synthesis of nanoparticles. These nanoparticles are often used as nanocarriers for therapeutic agents for healthcare applications. The best-known polysaccharides of plant origin include arabinogalactan, cellulose, guar gum, inulin, pectin, and starch.

4.2.1. Pectin

Pectin is a structural polysaccharide and consists of the main chain of D-galacturonic acid units linked by α-(1,4) glycosidic bonds and side chains of galactose, arabinose, and rhamnose. It is derived from various plant sources, mainly apples, citrus fruits, and sugar beets, but algae, such as Spirulina maxima can also produce it. It is water-soluble, biodegradable, biocompatible, and non-toxic, making it a promising biomaterial for the preparation of nanocarrier systems for various therapeutic agents for biomedical applications [197,198]. Pectin-based nanoparticles are unique systems for targeted drug delivery in the colon because pectin is readily degraded in the colon while remaining chemically intact in the stomach and small intestine [199].
Jacob et al. [200] synthesized polysaccharide nanoparticles based on pectin extracted from citrus fruits using the ionotropic gelation method based on the difference in their degree of esterification, namely high, low, and amidated low methoxylated pectin. Magnesium (Mg2+) was used as a divalent crosslinker. All three pectin samples proved to be potential delivery systems for hydrophobic drugs for oral administration. Bostanudin et al. [201] synthesized butylglyceryl-modified pectin nanoparticles by nanoprecipitation. The spherical nanoparticles produced were stable and less than 300 nm in diameter, making them appropriate drug delivery vehicles.

4.2.2. Starch

Starch is a natural biopolymer containing two different polysaccharide structures, amylose, and amylopectin. Amylose is a linear or slightly branched biopolymer and consists of glucose units linked by α-1-4 glycosidic bonds. Amylopectin is a highly branched biopolymer with short chains of glucose units linked by α-(1,4) glycosidic bonds and additional branches linked by α-1-6 glycosidic bonds. Like all other biopolymers, starch is biocompatible, biodegradable, nontoxic, nonimmunogenic, and renewable. Therefore, it represents a significant natural biopolymeric material for the preparation of nanoparticles as successful delivery systems for various drugs and bioactive substances with controlled release at the target site [202,203].
Farrag et al. [204] synthesized starch-based nanoparticles of various botanical origins into which the polyphenol quercetin was incorporated using the nanoprecipitation method. The results showed that the synthesized starch nanoparticles had a spherical shape. However, the origin of the starch had an influence on the size of the nanoparticles, the percentage of loading with quercetin, the release kinetics, and the antioxidant activity. Chin et al. [205] successfully synthesized paracetamol-loaded pH-responsive starch–citrate nanoparticles using a nanoprecipitation method. Synthesized nanocarriers are applicable for targeted drug delivery, with relatively high loading capacity and low toxicity. Chen et al. [206] demonstrated that starch-based nanoparticles could be used as nanocarriers for chemotherapeutic agents. In their study, they successfully prepared amphiphilic self-assembled glycyrrhetinic acid–biotin–starch nanoparticles with encapsulated hydrophobic anticancer drug doxorubicin. The nanoformulation showed enhanced cellular uptake and cytotoxic effect on HepG2 cells as a free drug.

4.2.3. Cellulose

Cellulose is a natural structural polysaccharide and the main component of many plant tissues. It is also found in seaweed and various non-pathogenic bacterial species. It is a linear biopolymer composed of D-glucose units linked by β-1,4-glycosidic bonds. Cellulose is the most abundant renewable biopolymer and is biocompatible, biodegradable, non-toxic, and stable under acidic conditions [95,126,207,208]. Cellulose-based nanomaterials (nanocellulose) can be used for a variety of biomedical applications, including wound healing, tissue engineering, drug delivery, bone regeneration, and as antibacterial and antifouling agents [209,210].
Chin et al. [211] demonstrated that curcumin-incorporated cellulose nanoparticles are a promising approach for curcumin delivery to the stomach and upper intestinal tract. In an in vitro study in simulated gastric fluid, higher stability and greater release of the drug were obtained. Cellulose-based nanoparticles are important for achieving target specificity and long-term efficacy in the delivery of therapeutic agents because they can be easily modified to obtain the appropriate chemical, mechanical, and physical properties [209].

4.2.4. Other Plant-Based Polysaccharide Nanoparticles

In addition to pectin and starch, there are other important biopolymers, including arabinogalactan, guar gum, and inulin, which can be used to produce nanoparticles as promising carriers for numerous therapeutics to improve their properties for healthcare applications. Most studies have been conducted primarily on the potential use of biopolymeric nanoparticles to deliver chemopreventive and chemotherapeutic agents [207,212,213]. For example, celecoxib has been successfully incorporated into guar gum nanoparticles to target colon cancer [214], as well as model drugs doxorubicin, 5-fluorouracil, rifampicin, rhodamine B, angiotensin II, antibiotic ciprofloxacin, and polyphenolic compounds, such as curcumin and caffeine. Furthermore, guar gum nanoparticles are suitable for oral as well as buccal, transdermal, and intravenous administration [215]. Inulin nanoparticles have also been developed for efficient delivery of the incorporated drug epirubicin [216]. In addition, inulin-based nanoparticles are potential carriers for unstable molecules in oral delivery systems. They ensure successful encapsulation, protection, and controlled release of quercetin for colon-targeted drug delivery [217]. Moreover, arabinogalactan-based nanoparticles are also suitable as carriers for various therapeutic agents, such as recombinant human thrombin [218].
Table 4 summarizes examples of biopolymeric nanoparticles of plant origin for use in various biomedical applications, including their preparation and the size of the synthesized nanoparticles.

5. Algae-Based Biopolymeric Nanoparticles Used for Healthcare Applications

Algae are autotrophs, mixotrophs, and heterotrophs and are found everywhere in freshwater sources and the marine environment. They are divided into microalgae and macroalgae, and both are important for the production of various biopolymers from which nanoparticles can be successfully synthesized [229].

5.1. Polysaccharide Nanoparticles of Algal Origin

Many polysaccharides, promising biopolymeric materials for biocompatible, biodegradable, and nontoxic nanoparticles, can also be obtained from algal species. Some important biopolymers of plant origin are also produced by various algae, such as cellulose and starch. Alginate and carrageenan are among the most commonly used biopolymers of algal origin.

5.1.1. Alginate

Alginate is a natural linear and unbranched polysaccharide consisting of a repeating unit of β-D-mannuronic acid (M-blocks) and α-L-guluronic acid (G-blocks) linked by 1,4-glycosidic bonds. The physicochemical properties of alginate are based on the composition, i.e., the M/G ratio and the length and sequence of the building blocks. The high content of M and G blocks contributes significantly to the thickening and gelling properties. Alginate is mainly derived from marine algae but can also be produced by some bacterial species. Due to its important properties, such as biodegradability, biocompatibility, non-toxicity, and non-immunogenicity, alginate is an important natural biomaterial for the preparation of nanoparticles for numerous therapeutic applications. Alginate nanoparticles successfully protect the incorporated therapeutic agents and ensure the controlled release of the therapeutic agent at the target site. The release properties of therapeutic agents are influenced by the presence of monomer units in alginate [127,230,231,232,233,234,235].
Spadari et al. [236] have successfully incorporated miltefosine, a drug with great antifungal potential, into alginate nanoparticles. These have been shown to be alternative nanocarriers for antifungal drugs to treat invasive fungal infections without having a cytotoxic effect on healthy cells, as may be the case when the free drug is administered. The bacteriocins of Lacticaseibacillus paracasei CNCM I-5369 show good activity against Gram-negative bacteria, including E. coli, which contains the mcr-1 gene that makes it resistant to the antibiotic colistin. In a study, Belguesmia et al. [237] successfully loaded alginate nanoparticles with bacteriocins, providing a successful therapeutic alternative in the fight against infections caused by Gram-negative bacteria. The results showed that antibacterial activity against E. coli increased significantly. Moreover, the alginate nanoparticles loaded with bacteriocins were not cytotoxic to human HT29 and animal IPEC-1 cell lines.

5.1.2. Carrageenan

Carrageenan is a linear sulfated polysaccharide obtained mainly from various species of seaweed. It is a hydrophilic biopolymer with a high molecular weight. Its polymer structure consists of 3-linked-β-D-galactopyranose and 4-linked-α-D-galactopyranose or 4-linked 3,6-anhydro-α-D-galactopyranose units. According to the structural features of the repeating disaccharide units, it can be classified into seven types, namely λ-, κ-, ι-, υ-, μ-, θ-, and ξ-carrageenan. Due to biodegradability, biocompatibility, bioavailability, non-toxicity, and thermostability, carrageenan is the ideal basis for the production of important nanoparticles based on natural polysaccharides in biomedical applications. It significantly contributes to improving the properties of incorporated therapeutic substances, increases bioavailability and durability, and prolongs the release of bioactive ingredients at the target site. In addition, carrageenan also has antioxidant, anticarcinogenic, anticoagulant, and immunomodulatory properties [61,238,239,240,241].
Fani et al. [242] used an electrospray method to synthesize carrageenan nanoparticles loaded with the sensitive bioactive compound D-limonene (R-(+)-limonene) and achieved an encapsulation efficiency of about 97%. Carrageenan-based nanoparticles were found to be effective nanocarrier systems for lipophilic compounds such as D-limonene, as both photostability and thermostability were significantly improved by encapsulation. Vijayakumar et al. [243] synthesized carrageenan-coated ZnO nanoparticles with successful antibacterial activity against MRSA. Moreover, the prepared nanoparticles had proven anti-inflammatory activity, good compatibility with human red blood cells, and were environmentally safe.
Table 5 summarizes examples of algae-based polysaccharide nanoparticles for various healthcare applications, including preparation methods and the size of produced nanoparticles.

6. Fungal-Based Biopolymeric Nanoparticles Used for Healthcare Applications

Fungi are an essential source of various natural compounds, including polysaccharides. Biodegradable and biocompatible nanoparticles, as carriers of drugs and other important therapeutic agents, can be synthesized from successfully extracted biopolymers from fungi by various methods.

Polysaccharide Nanoparticles of Fungal Origin

Pullulan is one of the most important and commonly used fungal polysaccharides for the preparation of biopolymeric nanoparticles. In addition to pullulan, the polysaccharide dextran, mainly of bacterial origin, and the protein chitosan, primarily derived from animals, can also be isolated from fungi.

Pullulan

Pullulan is a natural biopolymer obtained mainly by a fermentation process from the yeast-like fungus A. pullulans. The structure of pullulan contains maltotriose units linked by α-1,6 glycosidic bonds, which contributes significantly to the structural flexibility of pullulan. It is a nonionic linear exopolysaccharide easily soluble in water but insoluble in most organic solvents. Compared to other polysaccharides, the aqueous pullulan solution is stable and has a relatively low viscosity. Due to its advantageous properties, such as biodegradability, biocompatibility, non-immunogenicity, non-toxicity, non-mutagenicity, and non-carcinogenicity, pullulan is an important biomaterial for the preparation of nanoparticles based on natural polysaccharides, applicable for various biomedical use [61,111,249,250,251]. Pullulan-based nanoparticles are nanocarriers for various therapeutic substances, for example, antimicrobial agents and essential oils with inhibitory activity against pathogenic Gram-negative and Gram-positive bacteria and fungi, including multidrug-resistant and biofilm-forming bacteria [252].
Tao et al. [253] synthesized potential biopolymer-based nanocarriers to reduce the dose of chemotherapeutic drugs while increasing their delivery efficiency. They successfully incorporated the drug mitoxantrone into cholesterol-substituted pullulan nanoparticles, which showed successful growth inhibition of bladder cancer cells in vitro. Yuan et al. [254] synthesized cholesteryl-modified aminated pullulan nanoparticles loaded with cholesterol succinate, an acidic cholesterol ester. Sustained release of incorporated substance and cytotoxicity on Lewis lung cancer cells were obtained. In a study, Laha and Maiti [255] found that stearyl pullulan-based nanocarriers with the incorporated drug glipizide could prolong the release of the loaded drug under simulated gastrointestinal conditions. Thus, they proved to be favorable nanocarriers for poorly soluble drugs that can achieve controlled release and successfully improve the therapeutic efficiency of the incorporated drugs. Li et al. [256] demonstrated that pullulan–doxorubicin nanoparticles loaded with 1,1,2-trichlorotrifluoroethane are a new potential delivery system with a synergistic effect that significantly improves ablation and therapeutic efficacy.
Table 6 summarizes examples of polysaccharide nanoparticles of fungal origin for use in various biomedical applications, including synthesis methods and the size of nanoparticles produced.

7. Bacterial-Based Biopolymeric Nanoparticles Used for Healthcare Applications

Several bacterial species can produce various bioactive compounds, including polysaccharides, from which biodegradable, biocompatible, and nontoxic nanocarriers for potential use in biomedical applications can be prepared.

7.1. Polysaccharide Nanoparticles of Bacterial Origin

Polysaccharides that are successfully used to produce biopolymeric nanoparticles of bacterial origin include dextran, xanthan gum, levan, and gellan gum, of which dextran is the best known. In addition, pullulan can also be of bacterial origin. Polysaccharide-based nanoparticles of bacterial origin can successfully enhance bioavailability and consequently reduce the side effect of the incorporated drug or other therapeutic substance.

7.1.1. Dextran

Dextran is a natural, branched exopolysaccharide with a hydrophilic nature. It is produced mainly by some Gram-positive bacteria such as Leuconostoc and Streptococcus. The main dextran chain consists of glucose monomers linked by α-1,6 glycosidic bonds and short-side branches linked by α-1,3 glycosidic bonds. However, the properties of dextran, such as branching and molecular weight, may vary depending on the bacterial strain and production conditions. Due to its advantageous properties, such as biodegradability, biocompatibility, non-toxicity, and non-immunogenicity, it is considered as a suitable biomaterial for the preparation of polysaccharide nanoparticles as delivery systems for various therapeutic agents, including proteins and nucleic acids. Moreover, dextran nanoparticles successfully reduce the potential negative side effects of incorporated substances by delivering them to the target sites in a controlled manner [262,263,264].
In order to minimize the local and systemic side effects of local retinoblastoma chemotherapy, Delrish et al. [265] synthesized thiolated and methylated chitosan–carboxymethyl dextran nanoparticles by the ionic gelation method. They studied their biodistribution after intravitreal injection into the eyes of rats with retinoblastoma. The diameter of the prepared biocompatible polymer nanoparticles was 34 ± 3.78 nm and 42 ± 4.23 nm, respectively. These showed increased ocular bioavailability in retinoblastoma-induced rat eyes. In a study, Han et al. [266] found that paclitaxel-loaded dextran nanoparticles decorated with the RVG29 peptide can cross the blood–brain barrier and reach intracranial tumors. Therefore, they are suitable drug delivery systems for the treatment of C6 glioma. Jamwal et al. [267] synthesized a dextran-based insulin delivery system. An in vitro insulin release study was performed in an artificial gastric and artificial intestinal fluid and showed controlled release of insulin from synthesized glucose oxidase immobilized-acryloyl crosslinked dextran dialdehyde nanoparticles.

7.1.2. Other Bacterial-Based Polysaccharide Nanoparticles

Other bacterial-derived polysaccharides, such as xanthan gum, levan, and gellan gum, are also nontoxic, biodegradable, and biocompatible polymers for the synthesis of biopolymeric nanoparticles promising for biomedical applications as delivery systems. Therefore, various therapeutic substances have already been successfully incorporated into bacterial-based polysaccharide nanoparticles. For example, gellan gum and levan nanoparticles have been successfully loaded with resveratrol for oral administration [268,269], and the hydrophilic antihypertensive drug Atenol was incorporated into gellan gum nanoparticles [270]. Moreover, a chemotherapy drug, doxorubicin, was also successfully incorporated into xanthan gum nanoparticles [271].
Table 7 summarizes examples of polysaccharide nanoparticles of bacterial origin for use in various healthcare applications, including preparation methods and size of the synthesized nanoparticles.

8. Incorporation of Different Therapeutic Substances into Biopolymeric Nanoparticles for Healthcare Applications

Biopolymeric nanoparticles are applicable for clinical use as delivery systems for various drugs, genes, and other therapeutic agents and tissue engineering. This is due to their favorable properties, especially biodegradability, biocompatibility, non-toxicity, and low induction of immune responses [278,279]. Since they allow targeted delivery of incorporated drugs to target sites, preventing toxic effects on healthy cells, they can be loaded with a high concentration of drugs, allowing enhanced therapeutic effects due to sustained release. The loaded drug is successfully protected from metabolic processes and thus retains its bioactivity [3,84].
Free drugs require high dosages to achieve a pharmacological effect and are potentially toxic. In contrast, biodegradable biopolymer nanocarriers represent unique and effective systems for various healthcare applications due to their favorable properties and enhanced drug delivery in the human body. Moreover, they successfully reduce the potential risks and drawbacks of conventional therapies. There are several routes of administration by which therapeutic agents can be introduced into the human body using biopolymeric nanoparticles, namely the enteral (oral, ocular, nasal) or parenteral (subcutaneous, intravenous, intramuscular) route. However, the appropriate route of administration is influenced by the disease state, the desired therapeutic effect, and the bioavailability of the drug [278,280].

8.1. Incorporation and Delivery of Bioactive Compounds

Biologically active compounds, such as polyphenolic compounds, have antioxidant, anti-inflammatory, antimicrobial, antitumor, antiallergic, and other beneficial biological activities. However, they are susceptible to environmental conditions such as light, heat, and oxygen and are, therefore, chemically unstable. Furthermore, they are poorly soluble in water and are rapidly metabolized [49]. These limitations can be successfully overcome by incorporating bioactive compounds into nanocarriers, such as biopolymeric nanoparticles [195]. It has already been confirmed that encapsulation of phenolic compounds in nanoparticles significantly improves their stability, water solubility, and bioavailability, as well as their biological activity. Therefore, biopolymeric nanoparticles are suitable carriers for these compounds, as they can also reduce the required dose of bioactive compounds, further reducing potential side effects [49].
Many bioactive compounds also have good antioxidant properties. However, their potential use in the treatment of diseases caused by oxidative stress, including neurodegenerative diseases, is a significant concern because brain targeting is still a major challenge. Nanoformulations with incorporated antioxidants are promising delivery systems because they protect antioxidant molecules from degradation while allowing improved bioavailability and targeted delivery to specific organs, cells, or tissues [31].
The bioactive ingredient curcumin has many beneficial properties for human health. Therefore, many studies have already been conducted on its encapsulation in various biopolymeric nanoparticles to provide protection from rapid degradation and enhance its therapeutic effects. Das et al. [281] successfully incorporated curcumin into albumin-based nanoparticles, which showed enhanced cellular uptake and cytotoxicity in human lung carcinoma cells (A549) compared with the free compound. Hassani et al. [282] successfully encapsulated curcumin into sodium alginate–gum Arabic nanoparticles for enhanced antioxidant and drug-release properties of curcumin. The curcumin alginate–gum Arabic nanoparticles were prepared by an ionotropic gelation technique with slight modifications using calcium chloride as a crosslinker. Curcumin-loaded nanoparticles have significant therapeutic potential in preventing and treating solid malignancies. Compared with free curcumin, they showed enhanced anticancer activity against HepG2, HT29, A549, and MCF-7 cancer cells, as demonstrated by a cytotoxicity study using the MTT assay. Nogueira et al. [283] have also successfully incorporated curcumin into biopolymeric nanoparticles based on the polysaccharide carrageenan. High hydrophobicity may hinder the in vivo bioavailability of curcumin, reducing its therapeutic effect. Consequently, its incorporation into biodegradable nanoparticles is crucial. Curcumin-loaded carrageenan nanoparticles are a potentially effective and safe system for the treatment of bone-related diseases. Starch nanoparticles can also be used as nanocarriers for curcumin. Acevedo-Guevara et al. [222] used natural and acetylated starch from green bananas to prepare nanoparticles. Curcumin was successfully incorporated into both nanoformulations with an encapsulation efficiency greater than 80%. However, the acetylated starch nanoparticles were more successful because the acetylation of starch caused stronger hydrogen bond interaction between curcumin and the starch matrix, making the encapsulation more effective. In addition, improved controlled release of the polyphenolic substance from the acetylated starch nanoparticles was achieved. By incorporating curcumin into ovalbumin/κ-carrageenan nanoparticles, Xie et al. [284] demonstrated high antioxidant activity and stability in simulated gastric fluid and enhanced thermal and light stability of curcumin. Fan et al. [285] synthesized nanoparticles based on BSA and dextran for the preparation of curcumin delivery systems. The results of the study showed that the prepared nanoparticles loaded with curcumin were stable. Further, the antioxidant activity of curcumin in Caco-2 cells compared to the free bioactive compound was significantly improved. Liu et al. [286] synthesized zein nanoparticles with incorporated curcumin and stabilized dual-coating shell structure in combination with sodium caseinate (SC) and sodium alginate (SA). Zein nanoparticles stabilized with SC-SA significantly improved the photochemical stability and antioxidant activity of curcumin and enabled controlled release under simulated gastrointestinal conditions. The nanocarriers prepared in this way, therefore, have the potential to successfully deliver chemically unstable hydrophobic bioactive compounds. They can improve the water solubility, photochemical stability, and antioxidant activity of bioactive compounds such as curcumin. The same conclusions were reached by Yao et al. [287], who used zein nanoparticles loaded with curcumin and an alginate/gelatin biopolymer coating in their study. Biopolymeric materials can form stable coatings on the surface of zein and other nanoparticles made of natural polymers, which contributes to improved pH stability of nanoparticles and inhibition of particle aggregation during long-term storage [192,287]. Sorasitthiyanukarn et al. [288] successfully prepared chitosan/alginate nanoparticles loaded with curcumin diglutaric acid, a prodrug of curcumin with enhanced solubility and antinociceptive activity. These were more stable than free curcumin diglutaric acid under UV radiation and in simulated gastrointestinal environments. Additionally, they showed enhanced anticancer activity against Caco-2, HepG2, and human breast cancer cells (MDA-MB-231), as well as higher in vitro cell uptake in Caco-2 cells. Therefore, the authors claim that chitosan/alginate nanoparticles are a potential system for the oral delivery of curcumin diglutaric acid for cancer treatment. In a more recent study [289], the same authors synthesized chitosan/alginate nanoparticles loaded with curcumin diethyl disuccinate, an ester prodrug of curcumin. Compared with the free therapeutic substance, these showed improved stability, bioavailability, bioaccessibility, cellular uptake, and cytotoxicity against HepG2. In addition, chitosan-κ carrageenan nanoparticles with incorporated α-mangostin, a xanthone derivative isolated from the pericarp extract of mangosteen (Garcinia mangostana L.), are also promising delivery systems exhibit anticancer and excellent physicochemical properties [290]. They showed increased solubility, prolonged release, and cytotoxicity against MCF-7 cancer cells.
Rathore et al. [291] incorporated silymarin, a flavonoid, into collagen-based nanoparticles. Silymarin is a bioactive compound with hepatoprotective and neuroprotective properties isolated from milk thistle. The silymarin-loaded nanoformulation showed improved therapeutic efficacy of the flavonoid against acute ischemia/reperfusion injury. Moreover, a lower drug dose can achieve an appropriate neuroprotective effect by using nanoparticles. In addition, Roy and Rhim [292] incorporated a natural antioxidant flavonoid quercetin into chitosan-based nanoparticles, which exhibited good antioxidant and antimicrobial properties. Quercetin was also successfully loaded into zein nanoparticles stabilized with soluble soybean polysaccharide. This nanoformulation significantly improved the encapsulation efficiency and photochemical stability of quercetin [293]. Cinnamaldehyde is another natural compound belonging to flavonoids. Subhaswaraj et al. [182] prepared chitosan nanoparticles loaded with cinnamaldehyde, which showed significantly higher anti-quorum sensing activity and antibiofilm activity against Pseudomonas aeruginosa compared to free cinnamaldehyde. Moreover, slow and sustained release of cinnamaldehyde was achieved in an in vitro release study. Liang et al. [294] incorporated the polyphenolic compound tannic acid into synthesized zein/pectin nanoparticles with an average diameter of 166 nm and achieved an encapsulation efficiency of 89%. The results showed good antioxidant activity of the incorporated tannic acid, gradually released from the nanoparticles under simulated gastrointestinal conditions, especially in the small intestine.
Anacardic acid is a hydroxybenzoic acid with exceptional antimicrobial properties. Araujo et al. [295] successfully loaded anacardic acid into zein nanoparticles using a nanoprecipitation method. This method is considered an economical, environmentally friendly, low-energy, and reproducible technique. Nanoencapsulation of anacardic acid even increased antimicrobial activity against S. aureus, P. aeruginosa, and Candida auris. In another study, zein nanoparticles loaded with anacardic acid showed high bactericidal activity against S. mutans and potential antiplaque activity [296]. Bernal-Mercado et al. [297] incorporated carvacrol, a monoterpenoid phenol extracted from oregano and thyme essential oils, into chitosan nanoparticles, enhancing their antibacterial and antibiofilm properties against P. aeruginosa. Furthermore, Jain et al. [298] successfully loaded beta-carotene into zein nanoparticles prepared by a modified phase separation technique. They demonstrated enhanced cellular uptake, cytotoxicity, and improved oral biopharmaceutical performance of beta-carotene.

8.2. Incorporation and Delivery of Conventional Drugs

Selective therapeutics targeting diseased areas is a significant challenge in the treatment of many diseases, especially various forms of cancer [299]. Different anticancer drugs are poorly soluble in water and may have multiple toxic side effects, which can be successfully eliminated by incorporation into different nanocarriers [219]. Therefore, chemopreventive and chemotherapeutic agents can be successfully encapsulated into various biopolymeric nanoparticles. These offer improved stability and solubility of the drug, as well as a longer half-life in the blood due to protection from modifications caused by metabolic enzymes. In addition, drug encapsulation successfully reduces drug resistance and enables controlled drug release at the target site [31]. Nanoparticles have the ability to deliver incorporated drugs to the target organ, cells, or tissues, preventing the potentially toxic effects of drugs on healthy cells, as is the case with conventional forms of drug delivery. As a result, the amount of drug administered can also be reduced to achieve the desired therapeutic effect [300]. They are also potential gene nanocarriers due to their nanoscale size, high surface-to-volume ratio, and stability [31].
Yu et al. [219] successfully incorporated the anticancer drug maytansine with an encapsulation efficiency of 82.97% into zein nanoparticles prepared by phase separation to target the tumor and reduce the toxic side effects of the drug. Maytansinin-loaded zein nanoparticles have been shown to be non-toxic and promising nanocarriers for the treatment of non-small cell lung cancer, demonstrating enhanced antitumor efficacy in A549 cells compared to the free drug. Another anticancer drug, paclitaxel, was successfully incorporated into potato starch nanoparticles by Putro et al. [225]. The cytotoxicity assay results showed good biocompatibility of the nanoparticles with 7F2 mouse osteoblast cells. Abid et al. [299] synthesized enzyme-responsive dextran-based oligoester crosslinked nanoparticles containing the anticancer drug 5-fluorouracil as a potential oral delivery system to combat colon cancer. In a study by Ding et al. [224], 5-fluorouracil was also successfully incorporated into biopolymeric particles. The synthesized retrograded starch nanoparticles showed high absorption capacity and long in vitro release of the loaded drug, making them promising nanocarriers for colon-specific controlled release of 5-fluorouracil.
Pirfenidone is a drug with anti-inflammatory and antifibrotic activity used to treat idiopathic pulmonary fibrosis. Abnoos et al. [301] incorporated pirfenidone into chitosan/alginate nanoparticles with an encapsulation efficiency of 94%. An in vitro drug release study showed sustained release of the encapsulated drug, and skin penetration of the nanoparticle-loaded pirfenidone was compared to the free drug. Kianersi et al. [245] prepared alginate nanoparticles coated with chitosan and gelatin and loaded with the anti-inflammatory drug betamethasone sodium phosphate.
Thomas et al. [302] prepared starch-modified alginate nanoparticles using the ionotropic gelation method, into which they incorporated the drug theophylline and the protein BSA as model drugs. In vitro cytotoxicity studies showed that the nanoparticles were completely nontoxic and exhibited excellent biocompatibility. Nanoparticles prepared in this way successfully protected the incorporated drug in the acidic environment of the gastric fluid, and complete release of the drug in the intestinal fluid was achieved. Constantin et al. [303] prepared spherical nanoparticles based on a pullulan derivative incorporated with the model drug sodium diclofenac. In vitro studies revealed that the polymeric nanoparticles did not exhibit cytotoxicity at a pharmacologically relevant concentration of diclofenac and retained typical cell morphology.
Ursodeoxycholic acid is an oral drug for the treatment of primary biliary cirrhosis. Yu et al. [257] successfully demonstrated the neuroprotective effect of ursodeoxycholic acid-loaded pullulan acetate nanoparticles stabilized by poly(vinyl alcohol). Chinnaiyan et al. [221] prepared metformin-loaded pectin-based nanoparticles with an encapsulation efficiency of 68% by an ionic gelation method. The results showed reasonable stability in the presence of excess BSA and slow and sustained release of the drug at pH 6.8. In vivo stability of the nanoparticles and the possibility of prolonging the retention time of the drug in the bloodstream were demonstrated. Therefore, the pectin nanoparticles developed in this way are appropriate for enhancing the bioavailability of metformin for the successful treatment of type 2 diabetes.

8.3. Incorporation and Delivery of Antibiotics and Antimicrobial Agents

Due to increasing resistance to antibiotics and other antimicrobial agents, the search for new formulations for their administration has become increasingly important. Various metal nanoparticles are considered effective inhibitors of bacterial growth but are also highly toxic to human cells [304,305]. On the other hand, biocompatible and non-toxic biopolymeric nanoparticles also exhibit antimicrobial activity against various bacterial species [3]. Different nanoformulations, including polymeric nanoparticles in addition to liposomes, micelles, and solid lipid nanoparticles, can be considered suitable nanocarriers for antibiotics and other antimicrobial agents. Incorporating antibiotics into suitable nanoparticles can overcome antibiotic resistance, which increases the efficacy against pathogenic bacteria and contributes to the controlled release of the drug at the site of infection. In addition, encapsulation of antibiotics also has a positive effect on overcoming certain limitations, including poor solubility and stability of the drug and low permeation through biological barriers [306,307,308]. Thus, they can protect antibiotics from pH and/or enzymatic degradation [309].
Biopolymeric nanoparticles have been shown to be effective antibiotic carriers for combating antibiotic resistance. In vitro studies have demonstrated superior antibacterial activity against clinical isolates of multidrug-resistant E. coli, Klebsiella pneumoniae, and methicillin-resistant S. aureus (MRSA) compared to free antibiotics [306]. Furthermore, several antibiotics, such as colistin, can be toxic and provide low tissue penetration, so they are not commonly used. However, Scutera et al. [306] successfully incorporated the antibiotic colistin into HSA-based nanoparticles coated with chitosan. Compared to the free antibiotic, high antibacterial activity against the growth and inhibition of biofilm formation of multidrug-resistant Gram-negative bacterial species responsible for hospital-acquired severe infections (Acinetobacter baumannii and K. pneumoniae) was demonstrated. The antibacterial activity of the antibiotic imipenem and ciprofloxacin separately and in combination incorporated into albumin nanoparticles was increased, particularly against methicillin-resistant S. aureus and S. mutans [176]. Alginate/chitosan nanoparticles with incorporated antimicrobial drug tobramycin could be effectively used in the treatment of chronic infection with P. aeruginosa [310].
Furthermore, Liu et al. [311] prepared chitosan–sodium alginate nanoparticles into which they incorporated ε-polylysine, a homo-poly amino acid. In antimicrobial studies against the growth of E. coli, S. aureus, B. subtilis, and Micrococcus luteus, they showed three times higher bacteriostatic activity than free ε-polylysine. In vitro studies also showed prolonged release of the component from the nanoparticles. In patients with recurrent pulmonary infections, intrapulmonary administration of antibiotics may be beneficial compared with intravenous injection. Namely, inhalation can achieve a faster onset of action and a higher concentration of therapeutic agents in the lungs. Therefore, Falciani et al. [312] prepared a nanoformulation by binding the SET-M33 peptide, a not natural antimicrobial peptide, to single-chain dextran nanoparticles. Nanoparticles have been shown to be effective against pneumonia-causing P. aeruginosa in a mouse model by intrapulmonary administration. In addition, the retention time of the SET-M33 peptide in the lungs was effectively improved by incorporation into dextran-based nanoparticles. Moreover, Yu et al. [313] synthesized conjugates of chitosan nanoparticles and the antimicrobial peptide microcin J25. These have significantly enhanced antimicrobial activity against Gram-negative and Gram-positive bacteria while causing no toxicity in HEK293 T and Caco-2 cell lines.

8.4. Incorporation and Delivery of Extracts

It is known that natural extracts contain various biologically active substances that have a positive effect on health. Since they can be degraded in the gastrointestinal tract, their bioavailability is often poor. For the protection of sensitive compounds, the encapsulation of the extracts in different nanocarriers is crucial. A sustainable and suitable option are natural, non-toxic, biodegradable, and biocompatible protein- or polysaccharide-based nanoparticles to enhance their bioavailability and overcome biological barriers [246].
For this purpose, Costa et al. [246] successfully incorporated bioactive grape pomace extract into alginate and chitosan nanoparticles and investigated their potential as suitable nanocarriers for oral delivery. Alginate and chitosan nanoparticles protected the extract from the gastrointestinal environment, increasing the bioavailability of polyphenolic compounds in the extract and thus enhancing antioxidant and antimicrobial activity. In addition, the encapsulation of the extract also reduced its permeability through the intestinal barrier, allowing the extract to remain in the intestine longer, thus improving the prebiotic potential of the extract. Rahimivand et al. [314] successfully synthesized alginate nanocarriers with an incorporated ethanolic extract of Artemisia ciniformis, which increased cytotoxicity and induced apoptosis against AGS gastric cancer cells compared with the free extract. Encapsulation successfully protected the extracts from environmental stress. Wang et al. [223] synthesized starch nanoparticles obtained from ginkgo and corn, respectively, loaded with Ginkgo biloba extract using the nanoprecipitation method. Starch nanoparticles proved to be promising delivery systems for Ginko biloba extract or other bioactive compounds. The extract-loaded starch nanoparticles showed sustained release compared to the free extract in artificial gastric and intestinal juices. Additionally, Nallasamy et al. [226] have prepared starch-based nanoparticles into which they have successfully incorporated Triphala Churna extract, a polyherbal formulation rich in polyphenols and vitamin C that has rejuvenating and anti-aging effects. Encapsulation in biopolymer nanoparticles increased the stability, solubility, and efficacy of the Ayurvedic drug and achieved sustained release. In addition, antioxidant and anticholinesterase activities were maintained. A strong antimicrobial activity against Salmonella typhi, Shigella dysenteriae, and S. aureus, as well as anti-biofilm activity against ATCC MRSA 33591 and clinical strain N7 were demonstrated. Soltanzadeh et al. [183] synthesized spherical, physically stable chitosan nanoparticles with a diameter of 174–898 nm, into which pomegranate peel extract was successfully encapsulated. The study indicates promising nano-delivery systems for extracts and the resulting protection and controlled delivery of natural, sensitive substances with antioxidant and antimicrobial properties. Furthermore, Beconcini et al. [315] incorporated cherry extract into chitosan nanoparticles, successfully protecting the antioxidants contained in the extract from degradation in the gastrointestinal tract.

8.5. Incorporation and Delivery of Essential Oils

The labile and volatile compounds in essential oils can easily evaporate or decompose and are poorly soluble in water, affecting their antioxidant and antibacterial properties. Incorporating essential oils into nanoparticles, such as natural and biodegradable biopolymeric particles, successfully protects the essential oil components from degradation under adverse environmental conditions, extends shelf life, and contributes to controlled release [316,317].
Hadid et al. [316] successfully synthesized chitosan-based nanoparticles loaded with extracted clove bud essential oil, which exhibited better antioxidant and antimicrobial activity against Listeria monocytogenes and S. aureus compared to free essential oil. Hasheminejad et al. [180] also successfully incorporated clove essential oil into chitosan nanoparticles, which increased antifungal activity against A. niger. The in vitro release studies showed a controlled release of the essential oil over 56 days. By successfully encapsulating basil (Ocimum basilicum L.) essential oil in chitosan nanoparticles, Cai et al. [181] demonstrated enhanced antibacterial activity and strong antibiofilm activity against E. coli and S. aureus. Similarly, mandarin (Citrus reticulata L.) essential oil was successfully loaded into chitosan-based nanoparticles [318]. The nanoformulation successfully damaged the cell membrane of E. coli and S. aureus and inhibited biofilm formation and destroyed mature biofilms.
By incorporating essential oil from nettle leaves into chitosan nanoparticles, Bagheri et al. [319] achieved improved antioxidant activity compared to free essential oil and a more effective inhibitory effect on the growth of E. coli and S. aureus. Soltanzadeh et al. [184] incorporated the essential oil of lemongrass (Cymbopogon commutatus) into chitosan nanoparticles and demonstrated a time- and pH-dependent release of the essential oil in an in vitro release study. The chitosan nanoparticles also successfully retained the essential oils and preserved their bioactivity. Compared to free nanoparticles, nanoparticles with incorporated essential oil exhibited stronger antibacterial and antifungal activities. Furthermore, the essential oil of peppermint (Mentha piperita) and green tea (Camellia sinensis) [320] were also successfully loaded into chitosan nanoparticles, which exhibited enhanced thermal stability, antioxidant and antimicrobial activity. Additionally, cumin (Cuminum cyminum) seed essential oil was successfully encapsulated in chitosan nanoparticles [321]. Therefore, chitosan-based nanoparticles have emerged as a novel approach for the production of new pharmaceutical products with antimicrobial activity.
Furthermore, Yoncheva et al. [322] successfully encapsulated oregano essential oil in chitosan-alginate nanoparticles. The thermal stability of the oil, as well as antibacterial and antimetabolic activity compared to pure oil, was improved. In addition, an in vitro cytotoxicity test on human keratinocytes and an in vivo skin irritation test in an animal model demonstrated the safety profile of the oil-loaded nanoparticles. Merino et al. [220] incorporated Thymbra capitata essential oil into zein nanoparticles prepared by the self-assembly method and achieved an encapsulation efficiency of 77.8%. Compared with the essential oil in suspension, the essential oil-loaded nanoparticles exhibited enhanced antimicrobial activity. In addition, zein nanoparticles enable the controlled release of essential oils, making them beneficial nanomaterials for pharmaceutical and cosmetic purposes. Cai et al. [323] successfully incorporated tea tree essential oil into gliadin-based nanoparticles stabilized with gum Arabic.

8.6. Incorporation and Delivery of Different Therapeutic Substances: A Summary

Many studies have already been conducted testing both protein and polysaccharide nanoparticles of different origins as promising biodegradable and biocompatible delivery systems for drugs and other therapeutic agents. The aim of the studies was the improvement of bioavailability and therapeutic effect, as well as preventing possible adverse side effects of the loaded cargo.
Table 8 shows examples of drugs and other therapeutic agents incorporated into biopolymer nanoparticles, including their potential therapeutic applications.

9. Regulatory Aspects of Biopolymer-Based Nanoparticles

Biopolymers play a crucial role in major industrial sectors. Their sales are increasing, however, their demand for biomedical applications will grow rapidly in the coming years.
Various nanoparticles represent successful materials in numerous health applications, but there are still concerns about their safe use and long-term effects on human health. Therefore, risk management and regulatory issues remain [327]. Although biopolymeric nanoparticles are considered safe, biocompatible, biodegradable, and nontoxic, clinical studies on their safe use in drug delivery are urgently needed. Considering the many successful studies on biopolymeric nanoparticles with incorporated therapeutic agents, many companies are already developing suitable biopolymer-based nanoparticle formulations, some of which are already close to pre-clinical and clinical trials or are already in Phase 1. An example of a commercial formulation that is in Phase 1 is DE-310, which consists of a topoisomerase I inhibitor (exatecan mesylate) and a biodegradable carboxymethyl-dextran polyalcohol polymer covalently linked via a Gly-Gly-Phe-Gly peptidyl linker. In a Phase 1 clinical trial, it achieved complete remission in one of 27 patients with metastatic adenocarcinoma studied and partial remission in one patient with metastatic pancreatic cancer. In addition, it led to stabilization of disease progression in 14 patients. Delimotecan (MEN 4901/T-0128) is also a Phase 1 dextran-based nanoformulation with incorporated camptothecin (T-2513) for the treatment of solid tumors. The alginate and calcium gluconate-based formulation IK-5001 is also in Phase 1 for the prevention of ventricular remodeling and congestive heart failure. Another formulation is already in Phase 2, chitosan-based Milican, with incorporated holmium-166 for the treatment of small hepatocellular carcinoma [83].
On the other hand, some formulations containing biopolymeric nanoparticles are already successfully used and approved by the Food and Drug Administration (FDA) for various applications, including cancer treatment [52]. The first nanoformulation containing biopolymeric nanoparticles for the treatment of advanced metastatic pancreatic, breast, and non-small lung cancer is Abraxane, which the FDA has approved. Abraxane is a commercial formulation of the hydrophobic anticancer drug paclitaxel loaded into albumin-based nanoparticles. The nanoformulation is intended for intravenous administration and contains nanoparticles of 135 nm diameter [133]. Yuan et al. [328] investigated the efficacy and mechanism of two commercially available formulations to eliminate cancer stem cells in vitro and in vivo. In their study, they found that Abraxane, which contains paclitaxel-loaded albumin nanoparticles, improved the efficacy of paclitaxel in eliminating breast cancer stem cells over Taxol, a micelle formulation of paclitaxel [133].
However, the regulatory requirements for biopolymeric nanoparticles need greater attention and further research to ensure that commercial formulations can successfully overcome the challenges associated with stability and, more importantly, safety.

10. Conclusions

Research in the field of various nanoparticles as delivery systems for drugs and other therapeutic agents for healthcare applications is growing rapidly. However, synthetic nanoparticles can be toxic and have other unfavorable properties. Therefore, nanoparticles derived from natural materials such as biopolymers of animal, plant, algal, fungal, and bacterial origin are receiving increasing attention. Namely, they offer many advantages that are important factors for their potential use as successful and safe nanocarriers for drugs in the human body. Precisely because of their biocompatibility, biodegradability, nontoxicity, antioxidant properties (natural polysaccharides), and stability, research on the potential use of protein- and polysaccharide-based nanoparticles as favorable nanocarriers for therapeutic agents as new forms of treatment has significantly increased in the last decade. Furthermore, biopolymeric nanoparticles enable efficient encapsulation of many therapeutic agents, ensuring successful protection from degradation in the gastrointestinal tract, preventing potential toxic side effects of the loaded therapeutic agents, and ensuring controlled release at the target site, making them particularly promising for oral administration.
This review aims to generally outline the current state of research in the field of promising delivery of biopolymeric nanoparticles loaded with various therapeutic agents, both drugs and antibiotics, as well as natural extracts and bioactive compounds for biomedical applications. Biopolymeric nanoparticles successfully increase the bioavailability and improve the therapeutic effect not only of drugs, antibiotics, and other antimicrobial agents but also of extracts and essential oils, especially naturally isolated bioactive compounds. As far as natural compounds and their incorporation into biopolymeric nanoparticles are concerned, most studies have been conducted on curcumin and other bioactive compounds, which have thus shown promise as nanocarriers for various biomedical purposes. However, different encapsulated extracts and essential oils show significantly enhanced antioxidant and antimicrobial activity. Therefore, nanoparticles prepared from natural biopolymers with incorporated natural extracts and bioactive compounds are a promising alternative to various conventional therapies and therapeutic agents that cause toxic side effects and cytotoxicity to healthy cells. Moreover, they may not reach the target sites at all, leading to drug resistance in patients.
Based on the reviewed literature, studies on the use of polysaccharide nanoparticles for healthcare applications predominate. Chitosan is among the most studied biopolymers for the preparation of nanoparticles for the successful delivery of various therapeutic agents, as demonstrated by numerous in vitro studies. It is one of the most readily available and economical biopolymers and, therefore, the most suitable for commercial application. Nevertheless, further studies are urgently needed because chitosan nanoparticles, despite their excellent potential, are less recommended for therapeutic purposes due to the possible transmission of diseases from animals to humans by animal-based nanoparticles. It should be emphasized that preclinical studies are crucial for potential commercial use. Therefore, it is recommended to focus more on plant and microbial polysaccharide-based nanoparticles. Nowadays, polysaccharides can be largely obtained from algae, which are considered to be of less concern for healthcare use. Although there is a great tendency to cultivate algae for other applications such as food applications, fuels, etc., it is reasonable to direct the cultivation towards the extraction of polysaccharides for the synthesis of algae-based nanoparticles. However, further studies are critically needed to ensure the production of the most therapeutically successful, safe, sustainable, and biodegradable biopolymeric nanoparticles for healthcare applications.

Author Contributions

N.K. and M.L. conceived and designed the article. N.K. studied the literature and wrote the manuscript. M.P. reviewed and edited the article. M.L. and Ž.K. were responsible for the financial part of the project and reviewed the article. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Slovenian Research Agency (ARRS) within the frame of program P2-0046 (Separation Processes and Production Design), project No. J2-3037 (Bionanotechnology as a tool for stabilization and applications of bioactive substances from natural sources), project No. L2-4430 (Production, Isolation, and Formulation of Health Beneficial Substances from Helichrysum italicum for Applications in Cosmetic Industry), and young researcher ARRS fellowship contract number No. 2595/FKKT-2022.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Prasad, M.; Lambe, U.P.; Brar, B.; Shah, I.; Jyoti, M.; Ranjan, K.; Rao, R.; Kumar, S.; Mahant, S.; Khurana, S.K.; et al. Nanotherapeutics: An Insight into Healthcare and Multi-Dimensional Applications in Medical Sector of the Modern World. Biomed. Pharmacother. 2018, 97, 1521–1537. [Google Scholar] [CrossRef] [PubMed]
  2. Joudeh, N.; Linke, D. Nanoparticle Classification, Physicochemical Properties, Characterization, and Applications: A Comprehensive Review for Biologists. J. Nanobiotechnology 2022, 20, 262. [Google Scholar] [CrossRef] [PubMed]
  3. Vodyashkin, A.A.; Kezimana, P.; Vetcher, A.A.; Stanishevskiy, Y.M. Biopolymeric Nanoparticles–Multifunctional Materials of the Future. Polymers 2022, 14, 2287. [Google Scholar] [CrossRef] [PubMed]
  4. Ingle, A.P.; Gupta, I.; Duran, N.; Rai, M. Chapter 29—Nanotherapy: A next Generation Hallmark for Combating Cancer. In Nanostructures for Cancer Therapy; Ficai, A., Grumezescu, A.M., Eds.; Micro and Nano Technologies; Elsevier: Amsterdam, The Netherlands, 2017; pp. 811–830. ISBN 978-0-323-46144-3. [Google Scholar]
  5. Xiao, X.; Teng, F.; Shi, C.; Chen, J.; Wu, S.; Wang, B.; Meng, X.; Essiet Imeh, A.; Li, W. Polymeric Nanoparticles—Promising Carriers for Cancer Therapy. Front. Bioeng. Biotechnol. 2022, 10, 1024143. [Google Scholar] [CrossRef]
  6. Kolahalam, L.A.; Kasi Viswanath, I.V.; Diwakar, B.S.; Govindh, B.; Reddy, V.; Murthy, Y.L.N. Review on Nanomaterials: Synthesis and Applications. Mater. Today Proc. 2019, 18, 2182–2190. [Google Scholar] [CrossRef]
  7. Sajid, M. Nanomaterials: Types, Properties, Recent Advances, and Toxicity Concerns. Curr. Opin. Environ. Sci. Health. 2022, 25, 100319. [Google Scholar] [CrossRef]
  8. Ijaz, I.; Gilani, E.; Nazir, A.; Bukhari, A. Detail Review on Chemical, Physical and Green Synthesis, Classification, Characterizations and Applications of Nanoparticles. Green Chem. Lett. Rev. 2020, 13, 223–245. [Google Scholar] [CrossRef]
  9. Teleanu, D.M.; Chircov, C.; Grumezescu, A.M.; Teleanu, R.I. Neurotoxicity of Nanomaterials: An Up-to-Date Overview. Nanomaterials 2019, 9, 96. [Google Scholar] [CrossRef]
  10. Subhan, M.A.; Choudhury, K.P.; Neogi, N. Advances with Molecular Nanomaterials in Industrial Manufacturing Applications. Nanomanufacturing 2021, 1, 75–97. [Google Scholar] [CrossRef]
  11. Saha, S.; Bansal, S.; Khanuja, M. Chapter 2—Classification of Nanomaterials and Their Physical and Chemical Nature. In Nano-Enabled Agrochemicals in Agriculture; Ghorbanpour, M., Shahid, M.A., Eds.; Academic Press: Cambridge, MA, USA, 2022; pp. 7–34. ISBN 978-0-323-91009-5. [Google Scholar]
  12. Xiong, R.; Grant, A.M.; Ma, R.; Zhang, S.; Tsukruk, V.V. Naturally-Derived Biopolymer Nanocomposites: Interfacial Design, Properties and Emerging Applications. Mater. Sci. Eng. R Rep. 2018, 125, 1–41. [Google Scholar] [CrossRef]
  13. Alkaç, İ.M.; Çerçi, B.; Timuralp, C.; Şen, F. 2—Nanomaterials and Their Classification. In Nanomaterials for Direct Alcohol Fuel Cells; Şen, F., Ed.; Micro and Nano Technologies; Elsevier: Amsterdam, The Netherlands, 2021; pp. 17–33. ISBN 978-0-12-821713-9. [Google Scholar]
  14. Kang, H.; Rho, S.; Stiles, W.R.; Hu, S.; Baek, Y.; Hwang, D.W.; Kashiwagi, S.; Kim, M.S.; Choi, H.S. Size-Dependent EPR Effect of Polymeric Nanoparticles on Tumor Targeting. Adv. Healthc. Mater. 2020, 9, 1901223. [Google Scholar] [CrossRef]
  15. Distaso, M. Potential Contribution of Nanotechnology to the Circular Economy of Plastic Materials. Acta Innov. 2020, 37, 57–66. [Google Scholar] [CrossRef]
  16. Vassal, M.; Rebelo, S.; Pereira, M.D.L. Metal Oxide Nanoparticles: Evidence of Adverse Effects on the Male Reproductive System. Int. J. Mol. Sci. 2021, 22, 8061. [Google Scholar] [CrossRef]
  17. Patel, K.D.; Singh, R.K.; Kim, H.-W. Carbon-Based Nanomaterials as an Emerging Platform for Theranostics. Mater. Horiz. 2019, 6, 434–469. [Google Scholar] [CrossRef]
  18. Khan, I.; Saeed, K.; Khan, I. Nanoparticles: Properties, Applications and Toxicities. Arab. J. Chem. 2019, 12, 908–931. [Google Scholar] [CrossRef]
  19. Khalid, K.; Tan, X.; Mohd Zaid, H.F.; Tao, Y.; Lye Chew, C.; Chu, D.-T.; Lam, M.K.; Ho, Y.-C.; Lim, J.W.; Chin Wei, L. Advanced in Developmental Organic and Inorganic Nanomaterial: A Review. Bioengineered 2020, 11, 328–355. [Google Scholar] [CrossRef] [PubMed]
  20. Wang, H.; Zhou, Y.; Sun, Q.; Zhou, C.; Hu, S.; Lenahan, C.; Xu, W.; Deng, Y.; Li, G.; Tao, S. Update on Nanoparticle-Based Drug Delivery System for Anti-Inflammatory Treatment. Front. Bioeng. Biotechnol. 2021, 9, 630352. [Google Scholar] [CrossRef]
  21. Chandrakala, V.; Aruna, V.; Angajala, G. Review on Metal Nanoparticles as Nanocarriers: Current Challenges and Perspectives in Drug Delivery Systems. Emergent Mater. 2022, 5, 1593–1615. [Google Scholar] [CrossRef]
  22. Jamkhande, P.G.; Ghule, N.W.; Bamer, A.H.; Kalaskar, M.G. Metal Nanoparticles Synthesis: An Overview on Methods of Preparation, Advantages and Disadvantages, and Applications. J. Drug Deliv. Sci. Technol. 2019, 53, 101174. [Google Scholar] [CrossRef]
  23. Amini, E.; Azadfallah, M. In Situ Synthesis of Silver Nanoparticles on Fiber Matrix for Preparing Antibacterial Paper. Biointerface Res. Appl. Chem. 2018, 8, 3449–3456. [Google Scholar]
  24. Amini, E.; Valls, C.; Yousefi, H.; Roncero, M.B. Ionic Liquid/ZnO Assisted Preparation of High Barrier Cellulose Nanocomposite Films by In Situ Ring-Opening Polymerization of Lactide Monomers. J. Polym. Environ. 2023, 1–19. [Google Scholar] [CrossRef]
  25. Adepu, S.; Ramakrishna, S. Controlled Drug Delivery Systems: Current Status and Future Directions. Molecules 2021, 26, 5905. [Google Scholar] [CrossRef]
  26. Feitosa, R.C.; Geraldes, D.C.; Beraldo-de-Araújo, V.L.; Costa, J.S.R.; Oliveira-Nascimento, L. Pharmacokinetic Aspects of Nanoparticle-in-Matrix Drug Delivery Systems for Oral/Buccal Delivery. Front. Pharmacol. 2019, 10, 1057. [Google Scholar] [CrossRef]
  27. Leitgeb, M.; Knez, Ž.; Primožič, M. Sustainable Technologies for Liposome Preparation. J. Supercrit. Fluids 2020, 165, 104984. [Google Scholar] [CrossRef]
  28. Kučuk, N.; Primožič, M.; Knez, Ž.; Leitgeb, M. Exosomes Engineering and Their Roles as Therapy Delivery Tools, Therapeutic Targets, and Biomarkers. Int. J. Mol. Sci. 2021, 22, 9543. [Google Scholar] [CrossRef] [PubMed]
  29. Yetisgin, A.A.; Cetinel, S.; Zuvin, M.; Kosar, A.; Kutlu, O. Therapeutic Nanoparticles and Their Targeted Delivery Applications. Molecules 2020, 25, 2193. [Google Scholar] [CrossRef]
  30. Yoon, M.S.; Lee, Y.J.; Shin, H.J.; Park, C.-W.; Han, S.-B.; Jung, J.-K.; Kim, J.-S.; Shin, D.H. Recent Advances and Challenges in Controlling the Spatiotemporal Release of Combinatorial Anticancer Drugs from Nanoparticles. Pharmaceutics 2020, 12, 1156. [Google Scholar] [CrossRef] [PubMed]
  31. Harish, V.; Tewari, D.; Gaur, M.; Yadav, A.B.; Swaroop, S.; Bechelany, M.; Barhoum, A. Review on Nanoparticles and Nanostructured Materials: Bioimaging, Biosensing, Drug Delivery, Tissue Engineering, Antimicrobial, and Agro-Food Applications. Nanomaterials 2022, 12, 457. [Google Scholar] [CrossRef] [PubMed]
  32. Lu, X.-Y.; Wu, D.-C.; Li, Z.-J.; Chen, G.-Q. Chpater 7—Polymer Nanoparticles. In Progress in Molecular Biology and Translational Science; Villaverde, A., Ed.; Nanoparticles in Translational Science and Medicine; Academic Press: Cambridge, MA, USA, 2011; Volume 104, pp. 299–323. [Google Scholar]
  33. Guo, S.; Fu, D.; Utupova, A.; Sun, D.; Zhou, M.; Jin, Z.; Zhao, K. Applications of Polymer-Based Nanoparticles in Vaccine Field. Nanotechnol. Rev. 2019, 8, 143–155. [Google Scholar] [CrossRef]
  34. Noreen, S.; Ma, J.-X.; Saeed, M.; Pervaiz, F.; Hanif, M.F.; Ahmed, B.; Farooq, M.I.; Akram, F.; Safdar, M.; Madni, A.; et al. Natural Polysaccharide-Based Biodegradable Polymeric Platforms for Transdermal Drug Delivery System: A Critical Analysis. Drug. Deliv. Transl. Res. 2022, 12, 2649–2666. [Google Scholar] [CrossRef]
  35. Gowthaman, N.S.K.; Lim, H.N.; Sreeraj, T.R.; Amalraj, A.; Gopi, S. Chapter 15—Advantages of Biopolymers over Synthetic Polymers: Social, Economic, and Environmental Aspects. In Biopolymers and Their Industrial Applications; Thomas, S., Gopi, S., Amalraj, A., Eds.; Elsevier: Amsterdam, The Netherlands, 2021; pp. 351–372. ISBN 978-0-12-819240-5. [Google Scholar]
  36. Maghsoudi, S.; Taghavi Shahraki, B.; Rabiee, N.; Fatahi, Y.; Dinarvand, R.; Tavakolizadeh, M.; Ahmadi, S.; Rabiee, M.; Bagherzadeh, M.; Pourjavadi, A.; et al. Burgeoning Polymer Nano Blends for Improved Controlled Drug Release: A Review. Int. J. Nanomed. 2020, 15, 4363–4392. [Google Scholar] [CrossRef] [PubMed]
  37. Baranwal, J.; Barse, B.; Fais, A.; Delogu, G.L.; Kumar, A. Biopolymer: A Sustainable Material for Food and Medical Applications. Polymers 2022, 14, 983. [Google Scholar] [CrossRef]
  38. Lakshminarayanan, R.; Ye, E.; Young, D.J.; Li, Z.; Loh, X.J. Recent Advances in the Development of Antimicrobial Nanoparticles for Combating Resistant Pathogens. Adv. Healthc. Mater. 2018, 7, 1701400. [Google Scholar] [CrossRef]
  39. Muir, V.G.; Burdick, J.A. Chemically Modified Biopolymers for the Formation of Biomedical Hydrogels. Chem. Rev. 2021, 121, 10908–10949. [Google Scholar] [CrossRef]
  40. Biswas, M.C.; Jony, B.; Nandy, P.K.; Chowdhury, R.A.; Halder, S.; Kumar, D.; Ramakrishna, S.; Hassan, M.; Ahsan, M.A.; Hoque, M.E.; et al. Recent Advancement of Biopolymers and Their Potential Biomedical Applications. J. Polym. Environ. 2022, 30, 51–74. [Google Scholar] [CrossRef]
  41. Kaur, G.; Kumar, V.; Mishra, A.K.; Mishra, S. Chapter 12—Toxicological Effect of Biopolymers and Their Applications. In Bio-Based Nanomaterials; Mishra, A.K., Hussain, C.M., Eds.; Micro and Nano Technologies; Elsevier: Amsterdam, The Netherlands, 2022; pp. 265–284. ISBN 978-0-323-85148-0. [Google Scholar]
  42. Kumar, S.; Basumatary, I.B.; Mukherjee, A.; Dutta, J. An Overview of Natural Biopolymers in Food Packaging. In Biopolymer-Based Food Packaging; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2022; pp. 1–28. ISBN 978-1-119-70231-3. [Google Scholar]
  43. Kleber, M.; Reardon, P. Biopolymers and Macromolecules. In Encyclopedia of Geochemistry: A Comprehensive Reference Source on the Chemistry of the Earth; White, W.M., Ed.; Encyclopedia of Earth Sciences Series; Springer International Publishing: Cham, Switzerland, 2018; pp. 148–153. ISBN 978-3-319-39312-4. [Google Scholar]
  44. 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] [PubMed]
  45. Sun, X.; Wang, H.; Li, S.; Song, C.; Zhang, S.; Ren, J.; Udenigwe, C.C. Maillard-Type Protein–Polysaccharide Conjugates and Electrostatic Protein–Polysaccharide Complexes as Delivery Vehicles for Food Bioactive Ingredients: Formation, Types, and Applications. Gels 2022, 8, 135. [Google Scholar] [CrossRef] [PubMed]
  46. Bealer, E.J.; Onissema-Karimu, S.; Rivera-Galletti, A.; Francis, M.; Wilkowski, J.; Salas-de la Cruz, D.; Hu, X. Protein–Polysaccharide Composite Materials: Fabrication and Applications. Polymers 2020, 12, 464. [Google Scholar] [CrossRef]
  47. Pathak, N.; Singh, P.; Singh, P.K.; Sharma, S.; Singh, R.P.; Gupta, A.; Mishra, R.; Mishra, V.K.; Tripathi, M. Biopolymeric Nanoparticles Based Effective Delivery of Bioactive Compounds toward the Sustainable Development of Anticancerous Therapeutics. Front. Nutr. 2022, 9, 963413. [Google Scholar] [CrossRef] [PubMed]
  48. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.d.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano Based Drug Delivery Systems: Recent Developments and Future Prospects. J. Nanobiotechnology 2018, 16, 71. [Google Scholar] [CrossRef]
  49. Nishimoto-Sauceda, D.; Romero-Robles, L.E.; Antunes-Ricardo, M. Biopolymer Nanoparticles: A Strategy to Enhance Stability, Bioavailability, and Biological Effects of Phenolic Compounds as Functional Ingredients. J. Sci. Food Agric. 2022, 102, 41–52. [Google Scholar] [CrossRef] [PubMed]
  50. Pulingam, T.; Foroozandeh, P.; Chuah, J.-A.; Sudesh, K. Exploring Various Techniques for the Chemical and Biological Synthesis of Polymeric Nanoparticles. Nanomaterials 2022, 12, 576. [Google Scholar] [CrossRef]
  51. Meng, R.; Zhu, H.; Wang, Z.; Hao, S.; Wang, B. Preparation of Drug-Loaded Albumin Nanoparticles and Its Application in Cancer Therapy. J. Nanomater. 2022, 2022, e3052175. [Google Scholar] [CrossRef]
  52. Hong, S.; Choi, D.W.; Kim, H.N.; Park, C.G.; Lee, W.; Park, H.H. Protein-Based Nanoparticles as Drug Delivery Systems. Pharmaceutics 2020, 12, 604. [Google Scholar] [CrossRef]
  53. Bora, A.; Mishra, P. Casein and Ag Nanoparticles: Synthesis, Characterization, and Their Application in Biopolymer-Based Bilayer Film. J. Food Process. Preserv. 2019, 43, e14062. [Google Scholar] [CrossRef]
  54. Miao, Y.; Yang, T.; Yang, S.; Yang, M.; Mao, C. Protein Nanoparticles Directed Cancer Imaging and Therapy. Nano Converg. 2022, 9, 2. [Google Scholar] [CrossRef] [PubMed]
  55. Liang, T.; Zhang, Z.; Jing, P. Black Rice Anthocyanins Embedded in Self-Assembled Chitosan/Chondroitin Sulfate Nanoparticles Enhance Apoptosis in HCT-116 Cells. Food Chem. 2019, 301, 125280. [Google Scholar] [CrossRef]
  56. Jain, A.; Singh, S.K.; Arya, S.K.; Kundu, S.C.; Kapoor, S. Protein Nanoparticles: Promising Platforms for Drug Delivery Applications. ACS Biomater. Sci. Eng. 2018, 4, 3939–3961. [Google Scholar] [CrossRef] [PubMed]
  57. Lindemann, H.; Kühne, M.; Grune, C.; Warncke, P.; Hofmann, S.; Koschella, A.; Godmann, M.; Fischer, D.; Heinzel, T.; Heinze, T. Polysaccharide Nanoparticles Bearing HDAC Inhibitor as Nontoxic Nanocarrier for Drug Delivery. Macromol. Biosci. 2020, 20, 2000039. [Google Scholar] [CrossRef]
  58. Chopde, S.; Datir, R.; Deshmukh, G.; Dhotre, A.; Patil, M. Nanoparticle Formation by Nanospray Drying & Its Application in Nanoencapsulation of Food Bioactive Ingredients. J. Agric. Res. 2020, 2, 100085. [Google Scholar] [CrossRef]
  59. Asadi, M.; Salami, M.; Hajikhani, M.; Emam-Djomeh, Z.; Aghakhani, A.; Ghasemi, A. Electrospray Production of Curcumin-Walnut Protein Nanoparticles. Food Biophys. 2021, 16, 15–26. [Google Scholar] [CrossRef]
  60. Khalid, A.; Ahmed, N.; Qindeel, M.; Asad, M.I.; Khan, G.M.; ur.Rehman, A. Development of Novel Biopolymer-Based Nanoparticles Loaded Cream for Potential Treatment of Topical Fungal Infections. Drug Dev. Ind. Pharm 2021, 47, 1090–1099. [Google Scholar] [CrossRef]
  61. Plucinski, A.; Lyu, Z.; Schmidt, J.B.V.K. Polysaccharide Nanoparticles: From Fabrication to Applications. J. Mater. Chem. B 2021, 9, 7030–7062. [Google Scholar] [CrossRef]
  62. Pedroso-Santana, S.; Fleitas-Salazar, N. Ionotropic Gelation Method in the Synthesis of Nanoparticles/Microparticles for Biomedical Purposes. Polym. Int. 2020, 69, 443–447. [Google Scholar] [CrossRef]
  63. Kohli, K.; Mujtaba, A.; Malik, R.; Amin, S.; Alam, M.S.; Ali, A.; Barkat, M.A.; Ansari, M.J. Development of Natural Polysaccharide–Based Nanoparticles of Berberine to Enhance Oral Bioavailability: Formulation, Optimization, Ex Vivo, and In Vivo Assessment. Polymers 2021, 13, 3833. [Google Scholar] [CrossRef]
  64. Habibi, N.; Mauser, A.; Ko, Y.; Lahann, J. Protein Nanoparticles: Uniting the Power of Proteins with Engineering Design Approaches. Adv. Sci. 2022, 9, 2104012. [Google Scholar] [CrossRef] [PubMed]
  65. Aljabali, A.A.; Rezigue, M.; Alsharedeh, R.H.; Obeid, M.A.; Mishra, V.; Serrano-Aroca, Á.; El-Tanani, M.; Tambuwala, M.M. Protein-Based Nanomaterials: A New Tool for Targeted Drug Delivery. Ther. Deliv. 2022, 13, 321–338. [Google Scholar] [CrossRef]
  66. Zhang, Y.; Sun, T.; Jiang, C. Biomacromolecules as Carriers in Drug Delivery and Tissue Engineering. Acta Pharm. Sin. B. 2018, 8, 34–50. [Google Scholar] [CrossRef] [PubMed]
  67. Rizzuti, B. Nanomedicines Meet Disordered Proteins: A Shift from Traditional Materials and Concepts to Innovative Polymers. J. Pers. Med. 2022, 12, 1662. [Google Scholar] [CrossRef]
  68. Santofimia-Castaño, P.; Xia, Y.; Peng, L.; Velázquez-Campoy, A.; Abián, O.; Lan, W.; Lomberk, G.; Urrutia, R.; Rizzuti, B.; Soubeyran, P.; et al. Targeting the Stress-Induced Protein NUPR1 to Treat Pancreatic Adenocarcinoma. Cells 2019, 8, 1453. [Google Scholar] [CrossRef]
  69. Rizzuti, B.; Lan, W.; Santofimia-Castaño, P.; Zhou, Z.; Velázquez-Campoy, A.; Abián, O.; Peng, L.; Neira, J.L.; Xia, Y.; Iovanna, J.L. Design of Inhibitors of the Intrinsically Disordered Protein NUPR1: Balance between Drug Affinity and Target Function. Biomolecules 2021, 11, 1453. [Google Scholar] [CrossRef]
  70. Bonucci, A.; Palomino-Schätzlein, M.; Malo de Molina, P.; Arbe, A.; Pierattelli, R.; Rizzuti, B.; Iovanna, J.L.; Neira, J.L. Crowding Effects on the Structure and Dynamics of the Intrinsically Disordered Nuclear Chromatin Protein NUPR1. Front. Mol. Biosci. 2021, 8, 684622. [Google Scholar] [CrossRef] [PubMed]
  71. Karami, E.; Behdani, M.; Kazemi-Lomedasht, F. Albumin Nanoparticles as Nanocarriers for Drug Delivery: Focusing on Antibody and Nanobody Delivery and Albumin-Based Drugs. J. Drug Deliv. Sci. Technol. 2020, 55, 101471. [Google Scholar] [CrossRef]
  72. Carter, B.G.; Cheng, N.; Kapoor, R.; Meletharayil, G.H.; Drake, M.A. Invited Review: Microfiltration-Derived Casein and Whey Proteins from Milk. J. Dairy Sci. 2021, 104, 2465–2479. [Google Scholar] [CrossRef] [PubMed]
  73. Davison-Kotler, E.; Marshall, W.S.; García-Gareta, E. Sources of Collagen for Biomaterials in Skin Wound Healing. Bioengineering 2019, 6, 56. [Google Scholar] [CrossRef] [PubMed]
  74. Alipal, J.; Mohd Pu’ad, N.A.S.; Lee, T.C.; Nayan, N.H.M.; Sahari, N.; Basri, H.; Idris, M.I.; Abdullah, H.Z. A Review of Gelatin: Properties, Sources, Process, Applications, and Commercialisation. Mater. Today Proc. 2021, 42, 240–250. [Google Scholar] [CrossRef]
  75. Donato, R.K.; Mija, A. Keratin Associations with Synthetic, Biosynthetic and Natural Polymers: An Extensive Review. Polymers 2020, 12, 32. [Google Scholar] [CrossRef]
  76. Su, D.; Ding, S.; Shi, W.; Huang, X.; Jiang, L. Bombyx Mori Silk-Based Materials with Implication in Skin Repair: Sericin versus Regenerated Silk Fibroin. J. Biomater. Appl. 2019, 34, 36–46. [Google Scholar] [CrossRef]
  77. Mehanna, M.M.; Mneimneh, A.T. Updated but Not Outdated “Gliadin”: A Plant Protein in Advanced Pharmaceutical Nanotechnologies. Int. J. Pharm. 2020, 587, 119672. [Google Scholar] [CrossRef]
  78. Jaski, A.C.; Schmitz, F.; Horta, R.P.; Cadorin, L.; da Silva, B.J.G.; Andreaus, J.; Paes, M.C.D.; Riegel-Vidotti, I.C.; Zimmermann, L.M. Zein—A Plant-Based Material of Growing Importance: New Perspectives for Innovative Uses. Ind. Crops Prod. 2022, 186, 115250. [Google Scholar] [CrossRef]
  79. Shahbaz, A.; Hussain, N.; Basra, M.A.R.; Bilal, M. Polysaccharides-Based Nano-Hybrid Biomaterial Platforms for Tissue Engineering, Drug Delivery, and Food Packaging Applications. Starch Stärke 2022, 74, 2200023. [Google Scholar] [CrossRef]
  80. Sood, A.; Gupta, A.; Agrawal, G. Recent Advances in Polysaccharides Based Biomaterials for Drug Delivery and Tissue Engineering Applications. Carbohydr. Polym. Technol. Appl. 2021, 2, 100067. [Google Scholar] [CrossRef]
  81. Li, Z.; Lin, Z. Recent Advances in Polysaccharide-Based Hydrogels for Synthesis and Applications. Aggregate 2021, 2, e21. [Google Scholar] [CrossRef]
  82. Huh, M.S.; Lee, E.J.; Koo, H.; Yhee, J.Y.; Oh, K.S.; Son, S.; Lee, S.; Kim, S.H.; Kwon, I.C.; Kim, K. Polysaccharide-Based Nanoparticles for Gene Delivery. Top. Curr. Chem. 2017, 375, 31. [Google Scholar] [CrossRef]
  83. Miao, T.; Wang, J.; Zeng, Y.; Liu, G.; Chen, X. Polysaccharide-Based Controlled Release Systems for Therapeutics Delivery and Tissue Engineering: From Bench to Bedside. Adv. Sci. 2018, 5, 1700513. [Google Scholar] [CrossRef]
  84. Shokrani, H.; Shokrani, A.; Mohammad Sajadi, S.; Yazdi, M.K.; Seidi, F.; Jouyandeh, M.; Zarrintaj, P.; Kar, S.; Kim, S.-J.; Kuang, T.; et al. Polysaccharide-Based Nanocomposites for Biomedical Applications: A Critical Review. Nanoscale Horiz. 2022, 7, 1136–1160. [Google Scholar] [CrossRef]
  85. Sun, Y.; Jing, X.; Ma, X.; Feng, Y.; Hu, H. Versatile Types of Polysaccharide-Based Drug Delivery Systems: From Strategic Design to Cancer Therapy. Int. J. Mol. Sci. 2020, 21, 9159. [Google Scholar] [CrossRef]
  86. Khalid, A.; Ahmed, N.; Chaudhery, I.; Al-Jafary, M.A.; Al-Suhaimi, E.A.; Tarhini, M.; Lebaz, N.; Elaissari, A. Polysaccharide Chemistry in Drug Delivery, Endocrinology, and Vaccines. Chem. Eur. J. 2021, 27, 8437–8451. [Google Scholar] [CrossRef]
  87. Salave, S.; Rana, D.; Sharma, A.; Bharathi, K.; Gupta, R.; Khode, S.; Benival, D.; Kommineni, N. Polysaccharide Based Implantable Drug Delivery: Development Strategies, Regulatory Requirements, and Future Perspectives. Polysaccharides 2022, 3, 625–654. [Google Scholar] [CrossRef]
  88. Kou, S.G.; Peters, L.M.; Mucalo, M.R. Chitosan: A Review of Sources and Preparation Methods. Int. J. Biol. Macromol. 2021, 169, 85–94. [Google Scholar] [CrossRef]
  89. Reyes-Batlle, M.; Rodríguez-Talavera, I.; Sifaoui, I.; Rodríguez-Expósito, R.L.; Rocha-Cabrera, P.; Piñero, J.E.; Lorenzo-Morales, J. In vitro Amoebicidal Effects of Arabinogalactan-Based Ophthalmic Solution. Int. J. Parasitol. Drugs Drug Resist. 2021, 16, 9–16. [Google Scholar] [CrossRef]
  90. Farooq, A.; Patoary, M.K.; Zhang, M.; Mussana, H.; Li, M.; Naeem, M.A.; Mushtaq, M.; Farooq, A.; Liu, L. Cellulose from Sources to Nanocellulose and an Overview of Synthesis and Properties of Nanocellulose/Zinc Oxide Nanocomposite Materials. Int. J. Biol. Macromol. 2020, 154, 1050–1073. [Google Scholar] [CrossRef] [PubMed]
  91. Dehghani Soltani, M.; Meftahizadeh, H.; Barani, M.; Rahdar, A.; Hosseinikhah, S.M.; Hatami, M.; Ghorbanpour, M. Guar (Cyamopsis tetragonoloba L.) Plant Gum: From Biological Applications to Advanced Nanomedicine. Int. J. Biol. Macromol. 2021, 193, 1972–1985. [Google Scholar] [CrossRef] [PubMed]
  92. Sharma, G.; Sharma, S.; Kumar, A.; Al-Muhtaseb, A.H.; Naushad, M.; Ghfar, A.A.; Mola, G.T.; Stadler, F.J. Guar Gum and Its Composites as Potential Materials for Diverse Applications: A Review. Carbohydr. Polym. 2018, 199, 534–545. [Google Scholar] [CrossRef]
  93. Redondo-Cuenca, A.; Herrera-Vázquez, S.E.; Condezo-Hoyos, L.; Gómez-Ordóñez, E.; Rupérez, P. Inulin Extraction from Common Inulin-Containing Plant Sources. Ind. Crops Prod. 2021, 170, 113726. [Google Scholar] [CrossRef]
  94. Dranca, F.; Oroian, M. Extraction, Purification and Characterization of Pectin from Alternative Sources with Potential Technological Applications. Int. Food Res. J. 2018, 113, 327–350. [Google Scholar] [CrossRef]
  95. Seddiqi, H.; Oliaei, E.; Honarkar, H.; Jin, J.; Geonzon, L.C.; Bacabac, R.G.; Klein-Nulend, J. Cellulose and Its Derivatives: Towards Biomedical Applications. Cellulose 2021, 28, 1893–1931. [Google Scholar] [CrossRef]
  96. Khajouei, R.A.; Keramat, J.; Hamdami, N.; Ursu, A.-V.; Delattre, C.; Laroche, C.; Gardarin, C.; Lecerf, D.; Desbrières, J.; Djelveh, G.; et al. Extraction and Characterization of an Alginate from the Iranian Brown Seaweed Nizimuddinia zanardini. Int. J. Biol. Macromol. 2018, 118, 1073–1081. [Google Scholar] [CrossRef]
  97. Gomaa, M.; Fawzy, M.A.; Hifney, A.F.; Abdel-Gawad, K.M. Use of the Brown Seaweed Sargassum Latifolium in the Design of Alginate-Fucoidan Based Films with Natural Antioxidant Properties and Kinetic Modeling of Moisture Sorption and Polyphenolic Release. Food Hydrocoll. 2018, 82, 64–72. [Google Scholar] [CrossRef]
  98. Trica, B.; Delattre, C.; Gros, F.; Ursu, A.V.; Dobre, T.; Djelveh, G.; Michaud, P.; Oancea, F. Extraction and Characterization of Alginate from an Edible Brown Seaweed (Cystoseira barbata) Harvested in the Romanian Black Sea. Mar. Drugs 2019, 17, 405. [Google Scholar] [CrossRef]
  99. Montes, L.; Gisbert, M.; Hinojosa, I.; Sineiro, J.; Moreira, R. Impact of Drying on the Sodium Alginate Obtained after Polyphenols Ultrasound-Assisted Extraction from Ascophyllum Nodosum Seaweeds. Carbohydr. Polym. 2021, 272, 118455. [Google Scholar] [CrossRef] [PubMed]
  100. Bouasria, M.; El Mendili, Y.; Benzaama, M.-H.; Pralong, V.; Bardeau, J.-F.; Hennequart, F. Valorisation of Stranded Laminaria Digitata Seaweed as an Insulating Earth Material. Constr. Build. Mater. 2021, 308, 125068. [Google Scholar] [CrossRef]
  101. Cajnko, M.M.; Novak, U.; Likozar, B. Cascade Valorization Process of Brown Alga Seaweed Laminaria Hyperborea by Isolation of Polyphenols and Alginate. J. Appl. Phycol. 2019, 31, 3915–3924. [Google Scholar] [CrossRef]
  102. Li, S.-Y.; Wang, Z.-P.; Wang, L.-N.; Peng, J.-X.; Wang, Y.-N.; Han, Y.-T.; Zhao, S.-F. Combined Enzymatic Hydrolysis and Selective Fermentation for Green Production of Alginate Oligosaccharides from Laminaria Japonica. Bioresour. Technol. 2019, 281, 84–89. [Google Scholar] [CrossRef] [PubMed]
  103. Paul, S.; Salavarría, E.; Gil-Kodaka, P.; Villena, G.K. A de Novo Transcriptomic Approach to Study the Influence of Marine Water Depth in Macrocystis pyrifera Alginate Production. Aquat. Bot. 2020, 163, 103211. [Google Scholar] [CrossRef]
  104. Janarthanan, M.; Senthil Kumar, M. Extraction of Alginate from Brown Seaweeds and Evolution of Bioactive Alginate Film Coated Textile Fabrics for Wound Healing Application. J. Ind. Text. 2019, 49, 328–351. [Google Scholar] [CrossRef]
  105. Bahari, A.; Moelants, K.; Wallecan, J.; Mangiante, G.; Mazoyer, J.; Hendrickx, M.; Grauwet, T. Understanding the Effect of Time, Temperature and Salts on Carrageenan Extraction from Chondrus Crispus. Algal Res. 2021, 58, 102371. [Google Scholar] [CrossRef]
  106. Hughes, M.H.; Prado, H.J.; Rodríguez, M.C.; Michetti, K.; Leonardi, P.I.; Matulewicz, M.C. Carrageenans from Sarcothalia crispata and Gigartina skottsbergii: Structural Analysis and Interpolyelectrolyte Complex Formation for Drug Controlled Release. Mar. Biotechnol. 2018, 20, 706–717. [Google Scholar] [CrossRef]
  107. Naseri, A.; Jacobsen, C.; Sejberg, J.J.P.; Pedersen, T.E.; Larsen, J.; Hansen, K.M.; Holdt, S.L. Multi-Extraction and Quality of Protein and Carrageenan from Commercial Spinosum (Eucheuma denticulatum). Foods 2020, 9, 1072. [Google Scholar] [CrossRef]
  108. Das, A.K.; Sequeira, R.A.; Maity, T.K.; Prasad, K. Bio-Ionic Liquid Promoted Selective Coagulation of κ-Carrageenan from Kappaphycus Alvarezii Extract. Food Hydrocoll. 2021, 111, 106382. [Google Scholar] [CrossRef]
  109. Akrong, M.O.; Anning, A.K.; Addico, G.N.D.; Hogarh, J.N.; Adu-Gyamfi, A.; deGraft-Johnson, K.A.A.; Ale, M.; Meyer, A.S. Biomass and Carrageenan Yields of Hypnea Musciformis in Relation to Selected Environmental Variables in the Coastal Waters of Ghana. J. Appl. Phycol. 2022, 34, 2589–2601. [Google Scholar] [CrossRef]
  110. Haghighatpanah, N.; Mirzaee, H.; Khodaiyan, F.; Kennedy, J.F.; Aghakhani, A.; Hosseini, S.S.; Jahanbin, K. Optimization and Characterization of Pullulan Produced by a Newly Identified Strain of Aureobasidium Pullulans. Int. J. Biol. Macromol. 2020, 152, 305–313. [Google Scholar] [CrossRef] [PubMed]
  111. Raychaudhuri, R.; Naik, S.; Shreya, A.B.; Kandpal, N.; Pandey, A.; Kalthur, G.; Mutalik, S. Pullulan Based Stimuli Responsive and Sub Cellular Targeted Nanoplatforms for Biomedical Application: Synthesis, Nanoformulations and Toxicological Perspective. Int. J. Biol. Macromol. 2020, 161, 1189–1205. [Google Scholar] [CrossRef] [PubMed]
  112. Agrawal, S.; Budhwani, D.; Gurjar, P.; Telange, D.; Lambole, V. Pullulan Based Derivatives: Synthesis, Enhanced Physicochemical Properties, and Applications. Drug Deliv. 2022, 29, 3328–3339. [Google Scholar] [CrossRef]
  113. Abdulhameed, E.; Abdulsattar, J.; Majeed, H. Majeed Production, Characterization, and Antibacterial Effects of Dextran from Saccharomyces Cerevisiae Strains Obtained from Different Commercial Products Available in the Iraq Market. Int. J. Pharm. Res. 2020, 12, 2836–2844. [Google Scholar] [CrossRef]
  114. Sebastian, J.; Rouissi, T.; Brar, S.K.; Hegde, K.; Verma, M. Microwave-Assisted Extraction of Chitosan from Rhizopus Oryzae NRRL 1526 Biomass. Carbohydr. Polym. 2019, 219, 431–440. [Google Scholar] [CrossRef]
  115. Díaz-Montes, E. Dextran: Sources, Structures, and Properties. Polysaccharides 2021, 2, 554–565. [Google Scholar] [CrossRef]
  116. Rosca, I.; Petrovici, A.R.; Peptanariu, D.; Nicolescu, A.; Dodi, G.; Avadanei, M.; Ivanov, I.C.; Bostanaru, A.C.; Mares, M.; Ciolacu, D. Biosynthesis of Dextran by Weissella Confusa and Its In vitro Functional Characteristics. Int. J. Biol. Macromol. 2018, 107, 1765–1772. [Google Scholar] [CrossRef]
  117. Hu, Y.; Gänzle, M.G. Effect of Temperature on Production of Oligosaccharides and Dextran by Weissella Cibaria 10 M. Int. J. Food Microbiol. 2018, 280, 27–34. [Google Scholar] [CrossRef]
  118. Hamsan, M.H.; Shukur, M.F.; Aziz, S.B.; Kadir, M.F.Z. Dextran from Leuconostoc Mesenteroides-Doped Ammonium Salt-Based Green Polymer Electrolyte. Bull. Mater. Sci. 2019, 42, 57. [Google Scholar] [CrossRef]
  119. Jumma Kareem, A.; Abdul Sattar Salman, J. Production of Dextran from Locally Lactobacillus Spp. Isolates. Rep. Biochem. Mol. Biol. 2019, 8, 287–300. [Google Scholar] [PubMed]
  120. Dev, M.J.; Warke, R.G.; Warke, G.M.; Mahajan, G.B.; Patil, T.A.; Singhal, R.S. Advances in Fermentative Production, Purification, Characterization and Applications of Gellan Gum. Bioresour. Technol. 2022, 359, 127498. [Google Scholar] [CrossRef] [PubMed]
  121. Han, J.; Feng, H.; Wang, X.; Liu, Z.; Wu, Z. Levan from Leuconostoc Citreum BD1707: Production Optimization and Changes in Molecular Weight Distribution during Cultivation. BMC Biotechnol. 2021, 21, 14. [Google Scholar] [CrossRef] [PubMed]
  122. Vieira, A.M.; Zahed, F.; Crispim, A.C.; de Souza Bento, E.; França, R.d.F.O.; Pinheiro, I.O.; Pardo, L.A.; Carvalho, B.M. Production of Levan from Bacillus Subtilis Var. Natto and Apoptotic Effect on SH-SY5Y Neuroblastoma Cells. Carbohydr. Polym. 2021, 273, 118613. [Google Scholar] [CrossRef] [PubMed]
  123. Pei, F.; Ma, Y.; Chen, X.; Liu, H. Purification and Structural Characterization and Antioxidant Activity of Levan from Bacillus Megaterium PFY-147. Int. J. Biol. Macromol. 2020, 161, 1181–1188. [Google Scholar] [CrossRef]
  124. Kırtel, O.; Öner, E.T. Chapter 6: Levan Polysaccharide for Biomedical Applications. In Soft Matter for Biomedical Applications; Royal Society of Chemistry: London, UK, 2021; pp. 134–154. [Google Scholar]
  125. Sampaio, I.C.F.; Crugeira, P.J.L.; Soares, L.G.P.; dos Santos, J.N.; de Almeida, P.F.; Pinheiro, A.L.B.; Silveira, L. Composition of Xanthan Gum Produced by Xanthomonas Campestris Using Produced Water from a Carbonated Oil Field through Raman Spectroscopy. J. Photochem. Photobiol. B Biol. 2020, 213, 112052. [Google Scholar] [CrossRef]
  126. Kupnik, K.; Primožič, M.; Kokol, V.; Leitgeb, M. Nanocellulose in Drug Delivery and Antimicrobially Active Materials. Polymers 2020, 12, 2825. [Google Scholar] [CrossRef]
  127. Choukaife, H.; Doolaanea, A.A.; Alfatama, M. Alginate Nanoformulation: Influence of Process and Selected Variables. Pharmaceuticals 2020, 13, 335. [Google Scholar] [CrossRef]
  128. Hoogenboezem, E.N.; Duvall, C.L. Harnessing Albumin as a Carrier for Cancer Therapies. Adv. Drug Deliv. Rev. 2018, 130, 73–89. [Google Scholar] [CrossRef]
  129. Spada, A.; Emami, J.; Tuszynski, J.A.; Lavasanifar, A. The Uniqueness of Albumin as a Carrier in Nanodrug Delivery. Mol. Pharm. 2021, 18, 1862–1894. [Google Scholar] [CrossRef]
  130. Yan, F.; Li, H.; Zhong, Z.; Zhou, M.; Lin, Y.; Tang, C.; Li, C. Co-Delivery of Prednisolone and Curcumin in Human Serum Albumin Nanoparticles for Effective Treatment of Rheumatoid Arthritis. Int. J. Nanomed. 2019, 14, 9113–9125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Kuten Pella, O.; Hornyák, I.; Horváthy, D.; Fodor, E.; Nehrer, S.; Lacza, Z. Albumin as a Biomaterial and Therapeutic Agent in Regenerative Medicine. Int. J. Mol. Sci. 2022, 23, 10557. [Google Scholar] [CrossRef] [PubMed]
  132. Maurya, P.; Singh, S.; Mishra, N.; Pal, R.; Singh, N.; Parashar, P.; Saraf, S.A. Chapter 20—Albumin-Based Nanomaterials in Drug Delivery and Biomedical Applications. In Biopolymer-Based Nanomaterials in Drug Delivery and Biomedical Applications; Bera, H., Hossain, C.M., Saha, S., Eds.; Academic Press: Cambridge, MA, USA, 2021; pp. 465–496. ISBN 978-0-12-820874-8. [Google Scholar]
  133. Gawde, K.A.; Sau, S.; Tatiparti, K.; Kashaw, S.K.; Mehrmohammadi, M.; Azmi, A.S.; Iyer, A.K. Paclitaxel and Di-Fluorinated Curcumin Loaded in Albumin Nanoparticles for Targeted Synergistic Combination Therapy of Ovarian and Cervical Cancers. Colloids Surf. B Biointerfaces 2018, 167, 8–19. [Google Scholar] [CrossRef]
  134. Zhao, Y.; Cai, C.; Liu, M.; Zhao, Y.; Wu, Y.; Fan, Z.; Ding, Z.; Zhang, H.; Wang, Z.; Han, J. Drug-Binding Albumins Forming Stabilized Nanoparticles for Co-Delivery of Paclitaxel and Resveratrol: In vitro/in Vivo Evaluation and Binding Properties Investigation. Int. J. Biol. Macromol. 2020, 153, 873–882. [Google Scholar] [CrossRef] [PubMed]
  135. Ambrósio, J.A.R.; Pinto, B.C.D.S.; da Silva, B.G.M.; Passos, J.C.D.S.; Beltrame Junior, M.; Costa, M.S.; Simioni, A.R. BSA Nanoparticles Loaded-Methylene Blue for Photodynamic Antimicrobial Chemotherapy (PACT): Effect on Both Growth and Biofilm Formation by Candida Albicans. J. Biomater. Sci. Polym. Ed. 2020, 31, 2182–2198. [Google Scholar] [CrossRef] [PubMed]
  136. Arun, A.; Malrautu, P.; Laha, A.; Luo, H.; Ramakrishna, S. Collagen Nanoparticles in Drug Delivery Systems and Tissue Engineering. Appl. Sci. 2021, 11, 11369. [Google Scholar] [CrossRef]
  137. Xu, N.; Peng, X.-L.; Li, H.-R.; Liu, J.-X.; Cheng, J.-S.-Y.; Qi, X.-Y.; Ye, S.-J.; Gong, H.-L.; Zhao, X.-H.; Yu, J.; et al. Marine-Derived Collagen as Biomaterials for Human Health. Front. Nutr. 2021, 8, 702108. [Google Scholar] [CrossRef]
  138. Roată, C.-E.; Iacob, Ș.; Morărașu, Ș.; Livadaru, C.; Tudorancea, I.; Luncă, S.; Dimofte, M.-G. Collagen-Binding Nanoparticles: A Scoping Review of Methods and Outcomes. Crystals 2021, 11, 1396. [Google Scholar] [CrossRef]
  139. Kianfar, E. Protein Nanoparticles in Drug Delivery: Animal Protein, Plant Proteins and Protein Cages, Albumin Nanoparticles. J. Nanobiotechnology 2021, 19, 159. [Google Scholar] [CrossRef]
  140. Makkithaya, K.N.; Nadumane, S.; Zhuo, G.-Y.; Chakrabarty, S.; Mazumder, N.; Makkithaya, K.N.; Nadumane, S.; Zhuo, G.-Y.; Chakrabarty, S.; Mazumder, N. Nanoparticle Based Collagen Biomaterials for Wound Healing; IntechOpen: London, UK, 2022; ISBN 978-1-80355-412-9. [Google Scholar]
  141. Vijayakumar, S.; Vaseeharan, B. Antibiofilm, Anti Cancer and Ecotoxicity Properties of Collagen Based ZnO Nanoparticles. Adv. Powder Technol. 2018, 29, 2331–2345. [Google Scholar] [CrossRef]
  142. Ziaei Amiri, F.; Pashandi, Z.; Lotfibakhshaiesh, N.; Mirzaei-Parsa, M.J.; Ghanbari, H.; Faridi-Majidi, R. Cell Attachment Effects of Collagen Nanoparticles on Crosslinked Electrospun Nanofibers. Int. J. Artif. Organs 2021, 44, 199–207. [Google Scholar] [CrossRef]
  143. Lo, S.; Fauzi, M.B. Current Update of Collagen Nanomaterials—Fabrication, Characterisation and Its Applications: A Review. Pharmaceutics 2021, 13, 316. [Google Scholar] [CrossRef] [PubMed]
  144. Mondal, S.; Hoang, G.; Manivasagan, P.; Moorthy, M.S.; Vy Phan, T.T.; Kim, H.H.; Nguyen, T.P.; Oh, J. Rapid Microwave-Assisted Synthesis of Gold Loaded Hydroxyapatite Collagen Nano-Bio Materials for Drug Delivery and Tissue Engineering Application. Ceram. Int. 2019, 45, 2977–2988. [Google Scholar] [CrossRef]
  145. Gandhi, S.; Roy, I. Doxorubicin-Loaded Casein Nanoparticles for Drug Delivery: Preparation, Characterization and in vitro Evaluation. Int. J. Biol. Macromol. 2019, 121, 6–12. [Google Scholar] [CrossRef]
  146. Peñalva, R.; Morales, J.; González-Navarro, C.J.; Larrañeta, E.; Quincoces, G.; Peñuelas, I.; Irache, J.M. Increased Oral Bioavailability of Resveratrol by Its Encapsulation in Casein Nanoparticles. Int. J. Mol. Sci. 2018, 19, 2816. [Google Scholar] [CrossRef]
  147. Du, X.; Jing, H.; Wang, L.; Huang, X.; Mo, L.; Bai, X.; Wang, H. PH-Shifting Formation of Goat Milk Casein Nanoparticles from Insoluble Peptide Aggregates and Encapsulation of Curcumin for Enhanced Dispersibility and Bioactivity. LWT 2022, 154, 112753. [Google Scholar] [CrossRef]
  148. Peñalva, R.; Esparza, I.; Morales-Gracia, J.; González-Navarro, C.J.; Larrañeta, E.; Irache, J.M. Casein Nanoparticles in Combination with 2-Hydroxypropyl-β-Cyclodextrin Improves the Oral Bioavailability of Quercetin. Int. J. Pharm. 2019, 570, 118652. [Google Scholar] [CrossRef] [PubMed]
  149. Chen, L.; Wei, J.; An, M.; Zhang, L.; Lin, S.; Shu, G.; Yuan, Z.; Lin, J.; Peng, G.; Liang, X.; et al. Casein Nanoparticles as Oral Delivery Carriers of Mequindox for the Improved Bioavailability. Colloids Surf. B Biointerfaces 2020, 195, 111221. [Google Scholar] [CrossRef]
  150. Wang, Y.; Xue, Y.; Bi, Q.; Qin, D.; Du, Q.; Jin, P. Enhanced Antibacterial Activity of Eugenol-Entrapped Casein Nanoparticles Amended with Lysozyme against Gram-Positive Pathogens. Food Chem. 2021, 360, 130036. [Google Scholar] [CrossRef]
  151. Madkhali, O.; Mekhail, G.; Wettig, S.D. Modified Gelatin Nanoparticles for Gene Delivery. Int. J. Pharm. 2019, 554, 224–234. [Google Scholar] [CrossRef]
  152. Ferroni, C.; Varchi, G. Keratin-Based Nanoparticles as Drug Delivery Carriers. Appl. Sci. 2021, 11, 9417. [Google Scholar] [CrossRef]
  153. Gao, F.; Li, W.; Deng, J.; Kan, J.; Guo, T.; Wang, B.; Hao, S. Recombinant Human Hair Keratin Nanoparticles Accelerate Dermal Wound Healing. ACS Appl. Mater. Interfaces 2019, 11, 18681–18690. [Google Scholar] [CrossRef]
  154. Pham, D.T.; Tiyaboonchai, W. Fibroin Nanoparticles: A Promising Drug Delivery System. Drug Deliv. 2020, 27, 431–448. [Google Scholar] [CrossRef] [PubMed]
  155. Kumar Dan, A.; Aamna, B.; De, S.; Pereira-Silva, M.; Sahu, R.; Cláudia Paiva-Santos, A.; Parida, S. Sericin Nanoparticles: Future Nanocarrier for Target-Specific Delivery of Chemotherapeutic Drugs. J. Mol. Liq. 2022, 368, 120717. [Google Scholar] [CrossRef]
  156. Hu, Q.; Luo, Y. Recent Advances of Polysaccharide-Based Nanoparticles for Oral Insulin Delivery. Int. J. Biol. Macromol. 2018, 120, 775–782. [Google Scholar] [CrossRef] [PubMed]
  157. Chandrasekaran, M.; Kim, K.D.; Chun, S.C. Antibacterial Activity of Chitosan Nanoparticles: A Review. Processes 2020, 8, 1173. [Google Scholar] [CrossRef]
  158. Yanat, M.; Schroën, K. Preparation Methods and Applications of Chitosan Nanoparticles; with an Outlook toward Reinforcement of Biodegradable Packaging. React. Funct. Polym 2021, 161, 104849. [Google Scholar] [CrossRef]
  159. Herdiana, Y.; Wathoni, N.; Shamsuddin, S.; Joni, I.M.; Muchtaridi, M. Chitosan-Based Nanoparticles of Targeted Drug Delivery System in Breast Cancer Treatment. Polymers 2021, 13, 1717. [Google Scholar] [CrossRef]
  160. Aibani, N.; Rai, R.; Patel, P.; Cuddihy, G.; Wasan, E.K. Chitosan Nanoparticles at the Biological Interface: Implications for Drug Delivery. Pharmaceutics 2021, 13, 1686. [Google Scholar] [CrossRef] [PubMed]
  161. Madni, A.; Kousar, R.; Naeem, N.; Wahid, F. Recent Advancements in Applications of Chitosan-Based Biomaterials for Skin Tissue Engineering. J. Bioresour. Bioprod. 2021, 6, 11–25. [Google Scholar] [CrossRef]
  162. Križnik, L.; Vasić, K.; Knez, Ž.; Leitgeb, M. Hyper-Activation of ß-Galactosidase from Aspergillus Oryzae via Immobilization onto Amino-Silane and Chitosan Magnetic Maghemite Nanoparticles. J. Clean. Prod. 2018, 179, 225–234. [Google Scholar] [CrossRef]
  163. Hojnik Podrepšek, G.; Knez, Ž.; Leitgeb, M. Development of Chitosan Functionalized Magnetic Nanoparticles with Bioactive Compounds. Nanomaterials 2020, 10, 1913. [Google Scholar] [CrossRef]
  164. Saeed, R.M.; Dmour, I.; Taha, M.O. Stable Chitosan-Based Nanoparticles Using Polyphosphoric Acid or Hexametaphosphate for Tandem Ionotropic/Covalent Crosslinking and Subsequent Investigation as Novel Vehicles for Drug Delivery. Front. Bioeng. Biotechnol. 2020, 8, 4. [Google Scholar] [CrossRef] [PubMed]
  165. Shim, S.; Yoo, H.S. The Application of Mucoadhesive Chitosan Nanoparticles in Nasal Drug Delivery. Mar. Drugs 2020, 18, 605. [Google Scholar] [CrossRef] [PubMed]
  166. Rostami, E. Progresses in Targeted Drug Delivery Systems Using Chitosan Nanoparticles in Cancer Therapy: A Mini-Review. J. Drug Deliv. Sci. Technol. 2020, 58, 101813. [Google Scholar] [CrossRef]
  167. Ansari, M.M.; Ahmad, A.; Mishra, R.K.; Raza, S.S.; Khan, R. Zinc Gluconate-Loaded Chitosan Nanoparticles Reduce Severity of Collagen-Induced Arthritis in Wistar Rats. ACS Biomater. Sci. Eng. 2019, 5, 3380–3397. [Google Scholar] [CrossRef]
  168. Tzeyung, A.S.; Md, S.; Bhattamisra, S.K.; Madheswaran, T.; Alhakamy, N.A.; Aldawsari, H.M.; Radhakrishnan, A.K. Fabrication, Optimization, and Evaluation of Rotigotine-Loaded Chitosan Nanoparticles for Nose-To-Brain Delivery. Pharmaceutics 2019, 11, 26. [Google Scholar] [CrossRef]
  169. Yu, A.; Shi, H.; Liu, H.; Bao, Z.; Dai, M.; Lin, D.; Lin, D.; Xu, X.; Li, X.; Wang, Y. Mucoadhesive Dexamethasone-Glycol Chitosan Nanoparticles for Ophthalmic Drug Delivery. Int. J. Pharm. 2020, 575, 118943. [Google Scholar] [CrossRef] [PubMed]
  170. Sharkawy, A.; Barreiro, M.F.; Rodrigues, A.E. New Pickering Emulsions Stabilized with Chitosan/Collagen Peptides Nanoparticles: Synthesis, Characterization and Tracking of the Nanoparticles after Skin Application. Colloids Surf. A Physicochem. Eng. Asp. 2021, 616, 126327. [Google Scholar] [CrossRef]
  171. Ribeiro, E.F.; de Barros-Alexandrino, T.T.; Assis, O.B.G.; Junior, A.C.; Quiles, A.; Hernando, I.; Nicoletti, V.R. Chitosan and Crosslinked Chitosan Nanoparticles: Synthesis, Characterization and Their Role as Pickering Emulsifiers. Carbohydr. Polym. 2020, 250, 116878. [Google Scholar] [CrossRef] [PubMed]
  172. Babaee, M.; Garavand, F.; Rehman, A.; Jafarazadeh, S.; Amini, E.; Cacciotti, I. Biodegradability, Physical, Mechanical and Antimicrobial Attributes of Starch Nanocomposites Containing Chitosan Nanoparticles. Int. J. Biol. Macromol. 2022, 195, 49–58. [Google Scholar] [CrossRef]
  173. Fong, S.S.; Foo, Y.Y.; Saw, W.S.; Leo, B.F.; Teo, Y.Y.; Chung, I.; Goh, B.T.; Misran, M.; Imae, T.; Chang, C.-C.; et al. Chitosan-Coated-PLGA Nanoparticles Enhance the Antitumor and Antimigration Activity of Stattic—A STAT3 Dimerization Blocker. Int. J. Nanomed. 2022, 17, 137–150. [Google Scholar] [CrossRef]
  174. Qu, N.; Sun, Y.; Li, Y.; Hao, F.; Qiu, P.; Teng, L.; Xie, J.; Gao, Y. Docetaxel-Loaded Human Serum Albumin (HSA) Nanoparticles: Synthesis, Characterization, and Evaluation. Biomed. Eng. Online 2019, 18, 11. [Google Scholar] [CrossRef] [PubMed]
  175. Abolhassani, H.; Shojaosadati, S.A. A Comparative and Systematic Approach to Desolvation and Self-Assembly Methods for Synthesis of Piperine-Loaded Human Serum Albumin Nanoparticles. Colloids Surf. B Biointerfaces 2019, 184, 110534. [Google Scholar] [CrossRef]
  176. Shafiei, S.; Hassanshahian, M.; Shakeri, S.; Hamayeli, H. Evaluation the Antibacterial Activity of Nanoantibiotics Imipenem and Ciprofloxacin Loaded in Human Serum Albumin against Some Antibiotic-Resistant Pathogenic Bacteria. J. Exp. Nanosci. 2020, 15, 350–362. [Google Scholar] [CrossRef]
  177. Demirkurt, B.; Cakan-Akdogan, G.; Akdogan, Y. Preparation of Albumin Nanoparticles in Water-in-Ionic Liquid Microemulsions. J. Mol. Liq. 2019, 295, 111713. [Google Scholar] [CrossRef]
  178. Prajapati, R.; Garcia-Garrido, E.; Somoza, Á. Albumin-Based Nanoparticles for the Delivery of Doxorubicin in Breast Cancer. Cancers 2021, 13, 3011. [Google Scholar] [CrossRef] [PubMed]
  179. Tang, L.; Fu, L.; Zhu, Z.; Yang, Y.; Sun, B.; Shan, W.; Zhang, Z. Modified Mixed Nanomicelles with Collagen Peptides Enhanced Oral Absorption of Cucurbitacin B: Preparation and Evaluation. Drug Deliv. 2018, 25, 862–871. [Google Scholar] [CrossRef]
  180. Hasheminejad, N.; Khodaiyan, F.; Safari, M. Improving the Antifungal Activity of Clove Essential Oil Encapsulated by Chitosan Nanoparticles. Food Chem. 2019, 275, 113–122. [Google Scholar] [CrossRef]
  181. Cai, M.; Wang, Y.; Wang, R.; Li, M.; Zhang, W.; Yu, J.; Hua, R. Antibacterial and Antibiofilm Activities of Chitosan Nanoparticles Loaded with Ocimum Basilicum L. Essential Oil. Int. J. Biol. Macromol. 2022, 202, 122–129. [Google Scholar] [CrossRef] [PubMed]
  182. Subhaswaraj, P.; Barik, S.; Macha, C.; Chiranjeevi, P.V.; Siddhardha, B. Anti Quorum Sensing and Anti Biofilm Efficacy of Cinnamaldehyde Encapsulated Chitosan Nanoparticles against Pseudomonas Aeruginosa PAO1. LWT 2018, 97, 752–759. [Google Scholar] [CrossRef]
  183. Soltanzadeh, M.; Peighambardoust, S.H.; Ghanbarzadeh, B.; Mohammadi, M.; Lorenzo, J.M. Chitosan Nanoparticles as a Promising Nanomaterial for Encapsulation of Pomegranate (Punica Granatum L.) Peel Extract as a Natural Source of Antioxidants. Nanomaterials 2021, 11, 1439. [Google Scholar] [CrossRef] [PubMed]
  184. Soltanzadeh, M.; Peighambardoust, S.H.; Ghanbarzadeh, B.; Mohammadi, M.; Lorenzo, J.M. Chitosan Nanoparticles Encapsulating Lemongrass (Cymbopogon Commutatus) Essential Oil: Physicochemical, Structural, Antimicrobial and in-Vitro Release Properties. Int. J. Biol. Macromol. 2021, 192, 1084–1097. [Google Scholar] [CrossRef]
  185. Reddy, N.; Rapisarda, M. Properties and Applications of Nanoparticles from Plant Proteins. Materials 2021, 14, 3607. [Google Scholar] [CrossRef] [PubMed]
  186. Gomes, A.; Sobral, P.J.D.A. Plant Protein-Based Delivery Systems: An Emerging Approach for Increasing the Efficacy of Lipophilic Bioactive Compounds. Molecules 2022, 27, 60. [Google Scholar] [CrossRef] [PubMed]
  187. Zhang, X.; Zhang, Z.; Liang, H.; Li, J.; Wen, L.; Geng, F.; Li, B. Influence of Solvent Polarity of Ethonal/Water Binary Solvent on the Structural, Emulsifying, Interfacial Rheology Properties of Gliadin Nanoparticles. J. Mol. Liq. 2021, 344, 117976. [Google Scholar] [CrossRef]
  188. Dubey, A.; Ramteke, S.; Jain, D. Preparation and Characterization of Gliadin Nanoparticles for Brain Drug Delivery. Asian J. Pharm. Clin. Res. 2018, 11, 345–347. [Google Scholar] [CrossRef]
  189. Alqahtani, M.S.; Syed, R.; Alshehri, M. Size-Dependent Phagocytic Uptake and Immunogenicity of Gliadin Nanoparticles. Polymers 2020, 12, 2576. [Google Scholar] [CrossRef]
  190. Wu, W.; Kong, X.; Zhang, C.; Hua, Y.; Chen, Y.; Li, X. Fabrication and Characterization of Resveratrol-Loaded Gliadin Nanoparticles Stabilized by Gum Arabic and Chitosan Hydrochloride. LWT 2020, 129, 109532. [Google Scholar] [CrossRef]
  191. Sharif, N.; Golmakani, M.-T.; Hajjari, M.M. Integration of Physicochemical, Molecular Dynamics, and in vitro Evaluation of Electrosprayed γ-Oryzanol-Loaded Gliadin Nanoparticles. Food Chem. 2022, 395, 133589. [Google Scholar] [CrossRef]
  192. Abdelsalam, A.M.; Somaida, A.; Ayoub, A.M.; Alsharif, F.M.; Preis, E.; Wojcik, M.; Bakowsky, U. Surface-Tailored Zein Nanoparticles: Strategies and Applications. Pharmaceutics 2021, 13, 1354. [Google Scholar] [CrossRef]
  193. Zhang, H.; van Os, W.L.; Tian, X.; Zu, G.; Ribovski, L.; Bron, R.; Bussmann, J.; Kros, A.; Liu, Y.; Zuhorn, I.S. Development of Curcumin-Loaded Zein Nanoparticles for Transport across the Blood–Brain Barrier and Inhibition of Glioblastoma Cell Growth. Biomater. Sci. 2021, 9, 7092–7103. [Google Scholar] [CrossRef] [PubMed]
  194. Cardenas Turner, J.; Collins, G.; Blaber, E.A.; Almeida, E.A.C.; Arinzeh, T.L. Evaluating the Cytocompatibility and Differentiation of Bone Progenitors on Electrospun Zein Scaffolds. J. Tissue. Eng. Regen. Med. 2020, 14, 173–185. [Google Scholar] [CrossRef] [PubMed]
  195. Nunes, R.; Baião, A.; Monteiro, D.; das Neves, J.; Sarmento, B. Zein Nanoparticles as Low-Cost, Safe, and Effective Carriers to Improve the Oral Bioavailability of Resveratrol. Drug Deliv. Transl. Res. 2020, 10, 826–837. [Google Scholar] [CrossRef] [PubMed]
  196. Hadavi, M.; Hasannia, S.; Faghihi, S.; Mashayekhi, F.; Homazadeh, H.; Mostofi, S.B. Zein Nanoparticle as a Novel BMP6 Derived Peptide Carrier for Enhanced Osteogenic Differentiation of C2C12 Cells. Artif. Cells Nanomed. Biotechnol. 2018, 46, 559–567. [Google Scholar] [CrossRef] [PubMed]
  197. Federico, O.-D.; Lizeth, L.-E.C.; Guadalupe, F.-A.K.; Zuleth, G.-R.M.; Gabriel, H.G.-R.; Alfonso, S.-V.J.; Agustín, R.-C. 7—Pectin Nanoparticles: Fabrication and Uses. In Polysaccharide Nanoparticles; Venkatesan, J., Kim, S.-K., Anil, S.P.d.R., Eds.; Micro and Nano Technologies; Elsevier: Amsterdam, The Netherlands, 2022; pp. 163–201. ISBN 978-0-12-822351-2. [Google Scholar]
  198. Rajapaksha, D.C.; Edirisinghe, S.L.; Nikapitiya, C.; Dananjaya, S.H.S.; Kwun, H.-J.; Kim, C.-H.; Oh, C.; Kang, D.-H.; De Zoysa, M. Spirulina Maxima Derived Pectin Nanoparticles Enhance the Immunomodulation, Stress Tolerance, and Wound Healing in Zebrafish. Mar. Drugs 2020, 18, 556. [Google Scholar] [CrossRef]
  199. Khotimchenko, M. Pectin Polymers for Colon-Targeted Antitumor Drug Delivery. Int. J. Biol. Macromol. 2020, 158, 1110–1124. [Google Scholar] [CrossRef]
  200. Jacob, E.M.; Borah, A.; Jindal, A.; Pillai, S.C.; Yamamoto, Y.; Maekawa, T.; Kumar, D.N.S. Synthesis and Characterization of Citrus-Derived Pectin Nanoparticles Based on Their Degree of Esterification. J. Mater. Res. 2020, 35, 1514–1522. [Google Scholar] [CrossRef]
  201. Bostanudin, M.F.; Arafat, M.; Sarfraz, M.; Górecki, D.C.; Barbu, E. Butylglyceryl Pectin Nanoparticles: Synthesis, Formulation and Characterization. Polymers 2019, 11, 789. [Google Scholar] [CrossRef] [PubMed]
  202. Troncoso, O.P.; Torres, F.G. Non-Conventional Starch Nanoparticles for Drug Delivery Applications. Med. Devices Sens. 2020, 3, e10111. [Google Scholar] [CrossRef]
  203. Campelo, P.H.; Sant’Ana, A.S.; Pedrosa Silva Clerici, M.T. Starch Nanoparticles: Production Methods, Structure, and Properties for Food Applications. Curr. Opin. Food Sci. 2020, 33, 136–140. [Google Scholar] [CrossRef]
  204. Farrag, Y.; Ide, W.; Montero, B.; Rico, M.; Rodríguez-Llamazares, S.; Barral, L.; Bouza, R. Preparation of Starch Nanoparticles Loaded with Quercetin Using Nanoprecipitation Technique. Int. J. Biol. Macromol. 2018, 114, 426–433. [Google Scholar] [CrossRef]
  205. Chin, S.F.; Romainor, A.N.B.; Pang, S.C.; Lee, B.K.; Hwang, S.S. PH-Responsive Starch-Citrate Nanoparticles for Controlled Release of Paracetamol. Starch Stärke 2019, 71, 1800336. [Google Scholar] [CrossRef]
  206. Chen, K.; Zhang, S.; Wang, H.; Wang, X.; Zhang, Y.; Yu, L.; Ke, L.; Gong, R. Fabrication of Doxorubicin-Loaded Glycyrrhetinic Acid-Biotin-Starch Nanoparticles and Drug Delivery into HepG2 Cells In vitro. Starch Stärke 2019, 71, 1800031. [Google Scholar] [CrossRef]
  207. Khine, Y.Y.; Stenzel, M.H. Surface Modified Cellulose Nanomaterials: A Source of Non-Spherical Nanoparticles for Drug Delivery. Mater. Horiz. 2020, 7, 1727–1758. [Google Scholar] [CrossRef]
  208. Amini, E.; Valls, C.; Roncero, M.B. Ionic Liquid-Assisted Bioconversion of Lignocellulosic Biomass for the Development of Value-Added Products. J. Clean. Prod. 2021, 326, 129275. [Google Scholar] [CrossRef]
  209. Abdelhamid, H.N.; Mathew, A.P. Cellulose-Based Nanomaterials Advance Biomedicine: A Review. Int. J. Mol. Sci. 2022, 23, 5405. [Google Scholar] [CrossRef]
  210. Amini, E.; Azadfallah, M.; Layeghi, M.; Talaei-Hassanloui, R. Silver-Nanoparticle-Impregnated Cellulose Nanofiber Coating for Packaging Paper. Cellulose 2016, 23, 557–570. [Google Scholar] [CrossRef]
  211. Ching, Y.C.; Gunathilake, T.M.S.U.; Chuah, C.H.; Ching, K.Y.; Singh, R.; Liou, N.-S. Curcumin/Tween 20-Incorporated Cellulose Nanoparticles with Enhanced Curcumin Solubility for Nano-Drug Delivery: Characterization and in vitro Evaluation. Cellulose 2019, 26, 5467–5481. [Google Scholar] [CrossRef]
  212. Zhu, M.; Huan, S.; Liu, S.; Li, Z.; He, M.; Yang, G.; Liu, S.; McClements, D.J.; Rojas, O.J.; Bai, L. Recent Development in Food Emulsion Stabilized by Plant-Based Cellulose Nanoparticles. Curr. Opin. Colloid Interface Sci. 2021, 56, 101512. [Google Scholar] [CrossRef]
  213. Jiménez-Sánchez, M.; Pérez-Morales, R.; Goycoolea, F.M.; Mueller, M.; Praznik, W.; Loeppert, R.; Bermúdez-Morales, V.; Zavala-Padilla, G.; Ayala, M.; Olvera, C. Self-Assembled High Molecular Weight Inulin Nanoparticles: Enzymatic Synthesis, Physicochemical and Biological Properties. Carbohydr. Polym. 2019, 215, 160–169. [Google Scholar] [CrossRef] [PubMed]
  214. Chandel, D.; Uppal, S.; Mehta, S.K.; Shukla, G. Preparation and Characterization of Celecoxib Entrapped Guar Gum Nanoparticles Targeted for Oral Drug Delivery against Colon Cancer: An In-Vitro Study. J. Drug Deliv. Ther. 2020, 10, 14–21. [Google Scholar] [CrossRef]
  215. Verma, D.; Sharma, S.K. Recent Advances in Guar Gum Based Drug Delivery Systems and Their Administrative Routes. Int. J. Biol. Macromol. 2021, 181, 653–671. [Google Scholar] [CrossRef]
  216. Afinjuomo, F.; Abdella, S.; Youssef, S.H.; Song, Y.; Garg, S. Inulin and Its Application in Drug Delivery. Pharmaceuticals 2021, 14, 855. [Google Scholar] [CrossRef] [PubMed]
  217. Ayala-Fuentes, J.C.; Gallegos-Granados, M.Z.; Villarreal-Gómez, L.J.; Antunes-Ricardo, M.; Grande, D.; Chavez-Santoscoy, R.A. Optimization of the Synthesis of Natural Polymeric Nanoparticles of Inulin Loaded with Quercetin: Characterization and Cytotoxicity Effect. Pharmaceutics 2022, 14, 888. [Google Scholar] [CrossRef]
  218. Veverka, M.; Dubaj, T.; Gallovič, J.; Veverková, E.; Šimon, P.; Lokaj, J.; Jorík, V. Formulations of Staphylococcus Aureus Bacteriophage in Biodegradable Beta-Glucan and Arabinogalactan-Based Matrices. J. Drug Deliv. Sci. Technol. 2020, 59, 101909. [Google Scholar] [CrossRef]
  219. Yu, X.; Wu, H.; Hu, H.; Dong, Z.; Dang, Y.; Qi, Q.; Wang, Y.; Du, S.; Lu, Y. Zein Nanoparticles as Nontoxic Delivery System for Maytansine in the Treatment of Non-Small Cell Lung Cancer. Drug Deliv. 2020, 27, 100–109. [Google Scholar] [CrossRef]
  220. Merino, N.; Berdejo, D.; Bento, R.; Salman, H.; Lanz, M.; Maggi, F.; Sánchez-Gómez, S.; García-Gonzalo, D.; Pagán, R. Antimicrobial Efficacy of Thymbra capitata (L.) Cav. Essential Oil Loaded in Self-Assembled Zein Nanoparticles in Combination with Heat. Ind. Crops Prod. 2019, 133, 98–104. [Google Scholar] [CrossRef]
  221. Chinnaiyan, S.K.; Karthikeyan, D.; Gadela, V.R. Development and Characterization of Metformin Loaded Pectin Nanoparticles for T2 Diabetes Mellitus. Pharm. Nanotechnol. 2018, 6, 253–263. [Google Scholar] [CrossRef]
  222. Acevedo-Guevara, L.; Nieto-Suaza, L.; Sanchez, L.T.; Pinzon, M.I.; Villa, C.C. Development of Native and Modified Banana Starch Nanoparticles as Vehicles for Curcumin. Int. J. Biol. Macromol. 2018, 111, 498–504. [Google Scholar] [CrossRef]
  223. Wang, T.; Wu, C.; Fan, G.; Li, T.; Gong, H.; Cao, F. Ginkgo Biloba Extracts-Loaded Starch Nano-Spheres: Preparation, Characterization, and in vitro Release Kinetics. Int. J. Biol. Macromol. 2018, 106, 148–157. [Google Scholar] [CrossRef] [PubMed]
  224. Ding, Y.; Lin, Q.; Kan, J. Development and Characteristics Nanoscale Retrograded Starch as an Encapsulating Agent for Colon-Specific Drug Delivery. Colloids Surf. B: Biointerfaces 2018, 171, 656–667. [Google Scholar] [CrossRef] [PubMed]
  225. Putro, J.N.; Ismadji, S.; Gunarto, C.; Soetaredjo, F.E.; Ju, Y.H. A Study of Anionic, Cationic, and Nonionic Surfactants Modified Starch Nanoparticles for Hydrophobic Drug Loading and Release. J. Mol. Liq. 2020, 298, 112034. [Google Scholar] [CrossRef]
  226. Nallasamy, P.; Ramalingam, T.; Nooruddin, T.; Shanmuganathan, R.; Arivalagan, P.; Natarajan, S. Polyherbal Drug Loaded Starch Nanoparticles as Promising Drug Delivery System: Antimicrobial, Antibiofilm and Neuroprotective Studies. Process Biochem. 2020, 92, 355–364. [Google Scholar] [CrossRef]
  227. Chin, S.F.; Jimmy, F.B.; Pang, S.C. Size Controlled Fabrication of Cellulose Nanoparticles for Drug Delivery Applications. J. Drug Deliv. Sci. Technol. 2018, 43, 262–266. [Google Scholar] [CrossRef]
  228. Kim, W.-S.; Lee, J.-Y.; Singh, B.; Maharjan, S.; Hong, L.; Lee, S.-M.; Cui, L.-H.; Lee, K.-J.; Kim, G.; Yun, C.-H.; et al. A New Way of Producing Pediocin in Pediococcus Acidilactici through Intracellular Stimulation by Internalized Inulin Nanoparticles. Sci. Rep. 2018, 8, 5878. [Google Scholar] [CrossRef]
  229. Aswathi Mohan, A.; Robert Antony, A.; Greeshma, K.; Yun, J.-H.; Ramanan, R.; Kim, H.-S. Algal Biopolymers as Sustainable Resources for a Net-Zero Carbon Bioeconomy. Bioresour. Technol. 2022, 344, 126397. [Google Scholar] [CrossRef] [PubMed]
  230. Hasnain, M.S.; Nayak, A.K.; Kurakula, M.; Hoda, M.N. Chapter 6—Alginate Nanoparticles in Drug Delivery. In Alginates in Drug Delivery; Nayak, A.K., Hasnain, M.S., Eds.; Academic Press: Cambridge, MA, USA, 2020; pp. 129–152. ISBN 978-0-12-817640-5. [Google Scholar]
  231. Severino, P.; da Silva, C.F.; Andrade, L.N.; de Lima Oliveira, D.; Campos, J.; Souto, E.B. Alginate Nanoparticles for Drug Delivery and Targeting. Curr. Pharm. Des. 2019, 25, 1312–1334. [Google Scholar] [CrossRef]
  232. Dodero, A.; Alberti, S.; Gaggero, G.; Ferretti, M.; Botter, R.; Vicini, S.; Castellano, M. An Up-to-Date Review on Alginate Nanoparticles and Nanofibers for Biomedical and Pharmaceutical Applications. Adv. Mater. Interfaces 2021, 8, 2100809. [Google Scholar] [CrossRef]
  233. Li, S.; Zhang, H.; Chen, K.; Jin, M.; Vu, S.H.; Jung, S.; He, N.; Zheng, Z.; Lee, M.-S. Application of Chitosan/Alginate Nanoparticle in Oral Drug Delivery Systems: Prospects and Challenges. Drug Deliv. 2022, 29, 1142–1149. [Google Scholar] [CrossRef]
  234. Rostami, E. Recent Achievements in Sodium Alginate-Based Nanoparticles for Targeted Drug Delivery. Polym. Bull. 2022, 79, 6885–6904. [Google Scholar] [CrossRef]
  235. Al-Hatamleh, M.A.I.; Alshaer, W.; Hatmal, M.M.; Lambuk, L.; Ahmed, N.; Mustafa, M.Z.; Low, S.C.; Jaafar, J.; Ferji, K.; Six, J.-L.; et al. Applications of Alginate-Based Nanomaterials in Enhancing the Therapeutic Effects of Bee Products. Front. Mol. Biosci. 2022, 9, 865833. [Google Scholar] [CrossRef]
  236. Spadari, C.D.C.; Bastiani, F.W.M.D.S.; Lopes, L.B.; Ishida, K. Alginate Nanoparticles as Non-Toxic Delivery System for Miltefosine in the Treatment of Candidiasis and Cryptococcosis. Int. J. Nanomed. 2019, 14, 5187–5199. [Google Scholar] [CrossRef]
  237. Belguesmia, Y.; Hazime, N.; Kempf, I.; Boukherroub, R.; Drider, D. New Bacteriocins from Lacticaseibacillus paracasei CNCM I-5369 Adsorbed on Alginate Nanoparticles Are Very Active against Escherichia coli. Int. J. Mol. Sci. 2020, 21, 8654. [Google Scholar] [CrossRef]
  238. Dong, Y.; Wei, Z.; Xue, C. Recent Advances in Carrageenan-Based Delivery Systems for Bioactive Ingredients: A Review. Trends Food Sci. Technol. 2021, 112, 348–361. [Google Scholar] [CrossRef]
  239. BeMiller, J.N. 13—Carrageenans. In Carbohydrate Chemistry for Food Scientists, 3rd ed.; BeMiller, J.N., Ed.; Elsevier: Amsterdam, The Netherlands, 2019; pp. 279–291. ISBN 978-0-12-812069-9. [Google Scholar]
  240. Alba, K.; Kontogiorgos, V. Seaweed Polysaccharides (Agar, Alginate Carrageenan). In Encyclopedia of Food Chemistry; Melton, L., Shahidi, F., Varelis, P., Eds.; Academic Press: Oxford, UK, 2019; pp. 240–250. ISBN 978-0-12-814045-1. [Google Scholar]
  241. Yegappan, R.; Selvaprithiviraj, V.; Amirthalingam, S.; Jayakumar, R. Carrageenan Based Hydrogels for Drug Delivery, Tissue Engineering and Wound Healing. Carbohydr. Polym. 2018, 198, 385–400. [Google Scholar] [CrossRef]
  242. Fani, N.; Enayati, M.H.; Rostamabadi, H.; Falsafi, S.R. Encapsulation of Bioactives within Electrosprayed κ-Carrageenan Nanoparticles. Carbohydr. Polym. 2022, 294, 119761. [Google Scholar] [CrossRef]
  243. Vijayakumar, S.; Saravanakumar, K.; Malaikozhundan, B.; Divya, M.; Vaseeharan, B.; Durán-Lara, E.F.; Wang, M.-H. Biopolymer K-Carrageenan Wrapped ZnO Nanoparticles as Drug Delivery Vehicles for Anti MRSA Therapy. Int. J. Biol. Macromol. 2020, 144, 9–18. [Google Scholar] [CrossRef]
  244. Thomas, D.; KurienThomas, K.; Latha, M.S. Preparation and Evaluation of Alginate Nanoparticles Prepared by Green Method for Drug Delivery Applications. Int. J. Biol. Macromol. 2020, 154, 888–895. [Google Scholar] [CrossRef]
  245. Kianersi, S.; Solouk, A.; Saber-Samandari, S.; Keshel, S.H.; Pasbakhsh, P. Alginate Nanoparticles as Ocular Drug Delivery Carriers. J. Drug Deliv. Sci. Technol. 2021, 66, 102889. [Google Scholar] [CrossRef]
  246. Costa, J.R.; Xavier, M.; Amado, I.R.; Gonçalves, C.; Castro, P.M.; Tonon, R.V.; Cabral, L.M.C.; Pastrana, L.; Pintado, M.E. Polymeric Nanoparticles as Oral Delivery Systems for a Grape Pomace Extract towards the Improvement of Biological Activities. Mater. Sci. Eng. C. 2021, 119, 111551. [Google Scholar] [CrossRef]
  247. Alallam, B.; Altahhan, S.; Taher, M.; Mohd Nasir, M.H.; Doolaanea, A.A. Electrosprayed Alginate Nanoparticles as CRISPR Plasmid DNA Delivery Carrier: Preparation, Optimization, and Characterization. Pharmaceuticals 2020, 13, 158. [Google Scholar] [CrossRef]
  248. Huang, W.; Wang, L.; Wei, Y.; Cao, M.; Xie, H.; Wu, D. Fabrication of Lysozyme/κ-Carrageenan Complex Nanoparticles as a Novel Carrier to Enhance the Stability and in vitro Release of Curcumin. Int. J. Biol. Macromol. 2020, 146, 444–452. [Google Scholar] [CrossRef]
  249. Ganie, S.A.; Rather, L.J.; Li, Q. A Review on Anticancer Applications of Pullulan and Pullulan Derivative Nanoparticles. Carbohydr. Polym. Technol. Appl. 2021, 2, 100115. [Google Scholar] [CrossRef]
  250. Grigoras, A.G. Drug Delivery Systems Using Pullulan, a Biocompatible Polysaccharide Produced by Fungal Fermentation of Starch. Environ. Chem. Lett. 2019, 17, 1209–1223. [Google Scholar] [CrossRef]
  251. Singh, R.S.; Kaur, N.; Hassan, M.; Kennedy, J.F. Pullulan in Biomedical Research and Development—A Review. Int. J. Biol. Macromol. 2021, 166, 694–706. [Google Scholar] [CrossRef]
  252. Rai, M.; Wypij, M.; Ingle, A.P.; Trzcińska-Wencel, J.; Golińska, P. Emerging Trends in Pullulan-Based Antimicrobial Systems for Various Applications. Int. J. Mol. Sci. 2021, 22, 13596. [Google Scholar] [CrossRef]
  253. Tao, X.; Tao, T.; Wen, Y.; Yi, J.; He, L.; Huang, Z.; Nie, Y.; Yao, X.; Wang, Y.; He, C.; et al. Novel Delivery of Mitoxantrone with Hydrophobically Modified Pullulan Nanoparticles to Inhibit Bladder Cancer Cell and the Effect of Nano-Drug Size on Inhibition Efficiency. Nanoscale Res. Lett. 2018, 13, 345. [Google Scholar] [CrossRef]
  254. Yuan, H.; Zhong, W.; Wang, R.; Zhou, P.; Nie, Y.; Hu, W.; Tao, X.; Yang, P. Preparation of Cholesteryl-Modified Aminated Pullulan Nanoparticles to Evaluate Nanoparticle of Hydrophobic Degree on Drug Release and Cytotoxicity. J. Nanomater. 2020, 2020, e7171209. [Google Scholar] [CrossRef]
  255. Laha, B.; Maiti, S. Design of Core-Shell Stearyl Pullulan Nanostructures for Drug Delivery. Mater. Today Proc. 2019, 11, 620–627. [Google Scholar] [CrossRef]
  256. Li, H.; Yu, C.; Zhang, J.; Li, Q.; Qiao, H.; Wang, Z.; Zeng, D. PH-Sensitive Pullulan-Doxorubicin Nanoparticles Loaded with 1,1,2-Trichlorotrifluoroethane as a Novel Synergist for High Intensity Focused Ultrasound Mediated Tumor Ablation. Int. J. Pharm. 2019, 556, 226–235. [Google Scholar] [CrossRef]
  257. Yu, K.S.; Oh, J.Y.; Kim, M.C.; Kang, S.H.; Lee, N.S.; Han, S.-Y.; Ryu, K.H.; Jeong, Y.G.; Kim, D.K. Controlled Release of Ursodeoxycholic Acid from Pullulan Acetate Nanoparticles to Modulate Glutamate-Induced Excitotoxicity in PC-12 Cells. J. Nanomater. 2018, 2018, e7130450. [Google Scholar] [CrossRef]
  258. Pavaloiu, R.-D.; Sha’at, F.; Hlevca, C.; Sha’at, M.; Sevcenco, C.; Petrescu, M.; Eremia, M.; Moscovici, M. Preliminary Evaluation of Pullulan Nanoparticles Loaded with Valsartan. Chem. Proc. 2020, 3, 139. [Google Scholar] [CrossRef]
  259. Yang, W.; Zhang, Y.; Wang, J.; Li, H.; Yang, H. Glycyrrhetinic Acid-Cyclodextrin Grafted Pullulan Nanoparticles Loaded Doxorubicin as a Liver Targeted Delivery Carrier. Int. J. Biol. Macromol. 2022, 216, 789–798. [Google Scholar] [CrossRef]
  260. Wu, S.; Yang, X.; Yang, X. Methotrexate and 10-Hydroxycamptothecine Loaded Pullulan Nanoparticles with the Targeting Property for Efficient Cancer Therapy. Mater. Technol. 2022, 37, 2777–2784. [Google Scholar] [CrossRef]
  261. Bera, H.; Abosheasha, M.A.; Ito, Y.; Ueda, M. Hypoxia-Responsive Pullulan-Based Nanoparticles as Erlotinib Carriers. Int. J. Biol. Macromol. 2021, 191, 764–774. [Google Scholar] [CrossRef]
  262. Ukkund, S.J.; Alke, B.; Taqui, S.N.; Syed, U.T. 1—Dextran Nanoparticles: Preparation and Applications. In Polysaccharide Nanoparticles; Venkatesan, J., Kim, S.-K., Anil, S., P. d, R., Eds.; Micro and Nano Technologies; Elsevier: Amsterdam, The Netherlands, 2022; pp. 1–31. ISBN 978-0-12-822351-2. [Google Scholar]
  263. Huang, G.; Huang, H. Application of Dextran as Nanoscale Drug Carriers. Nanomedicine 2018, 13, 3149–3158. [Google Scholar] [CrossRef]
  264. Chen, F.; Huang, G.; Huang, H. Preparation and Application of Dextran and Its Derivatives as Carriers. Int. J. Biol. Macromol. 2020, 145, 827–834. [Google Scholar] [CrossRef]
  265. Delrish, E.; Ghassemi, F.; Jabbarvand, M.; Lashay, A.; Atyabi, F.; Soleimani, M.; Dinarvand, R. Biodistribution of Cy5-Labeled Thiolated and Methylated Chitosan-Carboxymethyl Dextran Nanoparticles in an Animal Model of Retinoblastoma. J. Ophthalmic. Vis. Res. 2022, 17, 58–68. [Google Scholar] [CrossRef]
  266. Han, H.; Zhang, Y.; Jin, S.; Chen, P.; Liu, S.; Xie, Z.; Jing, X.; Wang, Z. Paclitaxel-Loaded Dextran Nanoparticles Decorated with RVG29 Peptide for Targeted Chemotherapy of Glioma: An in Vivo Study. New J. Chem. 2020, 44, 5692–5701. [Google Scholar] [CrossRef]
  267. Jamwal, S.; Ram, B.; Ranote, S.; Dharela, R.; Chauhan, G.S. New Glucose Oxidase-Immobilized Stimuli-Responsive Dextran Nanoparticles for Insulin Delivery. Int. J. Biol. Macromol. 2019, 123, 968–978. [Google Scholar] [CrossRef]
  268. Prezotti, F.G.; Boni, F.I.; Ferreira, N.N.; Silva, D.S.; Almeida, A.; Vasconcelos, T.; Sarmento, B.; Gremião, M.P.D.; Cury, B.S.F. Oral Nanoparticles Based on Gellan Gum/Pectin for Colon-Targeted Delivery of Resveratrol. Drug Dev. Ind. Pharm. 2020, 46, 236–245. [Google Scholar] [CrossRef]
  269. Cinan, E.; Cesur, S.; Erginer Haskoylu, M.; Gunduz, O.; Toksoy Oner, E. Resveratrol-Loaded Levan Nanoparticles Produced by Electrohydrodynamic Atomization Technique. Nanomaterials 2021, 11, 2582. [Google Scholar] [CrossRef]
  270. Sharma, R.; Sharma, U. Formulation and Characterization of Atenolol-Loaded Gellan Gum Nanoparticles. Indian J. Pharm. Sci. 2021, 83, 60–65. [Google Scholar] [CrossRef]
  271. Feng, Z.; Xu, J.; Ni, C. Preparation of Redox Responsive Modified Xanthan Gum Nanoparticles and the Drug Controlled Release. Int. J. Polym. Mater. Polym. Biomater. 2021, 70, 994–1001. [Google Scholar] [CrossRef]
  272. Kadota, K.; Yanagawa, Y.; Tachikawa, T.; Deki, Y.; Uchiyama, H.; Shirakawa, Y.; Tozuka, Y. Development of Porous Particles Using Dextran as an Excipient for Enhanced Deep Lung Delivery of Rifampicin. Int. J. Pharm. 2019, 555, 280–290. [Google Scholar] [CrossRef]
  273. Butzbach, K.; Konhäuser, M.; Fach, M.; Bamberger, D.N.; Breitenbach, B.; Epe, B.; Wich, P.R. Receptor-Mediated Uptake of Folic Acid-Functionalized Dextran Nanoparticles for Applications in Photodynamic Therapy. Polymers 2019, 11, 896. [Google Scholar] [CrossRef]
  274. Lee, S.; Stubelius, A.; Hamelmann, N.; Tran, V.; Almutairi, A. Inflammation-Responsive Drug-Conjugated Dextran Nanoparticles Enhance Anti-Inflammatory Drug Efficacy. ACS Appl. Mater. Interfaces 2018, 10, 40378–40387. [Google Scholar] [CrossRef]
  275. Kashyap, A.; Kaur, R.; Baldi, A.; Jain, U.K.; Chandra, R.; Madan, J. Chloroquine Diphosphate Bearing Dextran Nanoparticles Augmented Drug Delivery and Overwhelmed Drug Resistance in Plasmodium Falciparum Parasites. Int. J. Biol. Macromol. 2018, 114, 161–168. [Google Scholar] [CrossRef]
  276. Madkhali, O.A.; Sivagurunathan Moni, S.; Sultan, M.H.; Bukhary, H.A.; Ghazwani, M.; Alhakamy, N.A.; Meraya, A.M.; Alshahrani, S.; Alqahtani, S.S.; Bakkari, M.A.; et al. Formulation and Evaluation of Injectable Dextran Sulfate Sodium Nanoparticles as a Potent Antibacterial Agent. Sci. Rep. 2021, 11, 9914. [Google Scholar] [CrossRef]
  277. Muhammad, D.R.A.; Sedaghat Doost, A.; Gupta, V.; bin Sintang, M.D.; Van de Walle, D.; Van der Meeren, P.; Dewettinck, K. Stability and Functionality of Xanthan Gum–Shellac Nanoparticles for the Encapsulation of Cinnamon Bark Extract. Food Hydrocoll. 2020, 100, 105377. [Google Scholar] [CrossRef]
  278. Verma, M.L.; Dhanya, B.S.; Rani, V.; Thakur, M.; Jeslin, J.; Kushwaha, R. Carbohydrate and Protein Based Biopolymeric Nanoparticles: Current Status and Biotechnological Applications. Int. J. Biol. Macromol. 2020, 154, 390–412. [Google Scholar] [CrossRef] [PubMed]
  279. Ramachandraiah, K.; Hong, G.-P. Polymer Based Nanomaterials for Strategic Applications in Animal Food Value Chains. Food Rev. Int. 2022, 38, 1577–1606. [Google Scholar] [CrossRef]
  280. Jain, K.K. An Overview of Drug Delivery Systems. In Drug Delivery Systems; Jain, K.K., Ed.; Methods in Molecular Biology; Springer: New York, NY, USA, 2020; pp. 1–54. ISBN 978-1-4939-9798-5. [Google Scholar]
  281. Das, R.P.; Gandhi, V.V.; Singh, B.G.; Kunwar, A.; Kumar, N.N.; Priyadarsini, K.I. Preparation of Albumin Nanoparticles: Optimum Size for Cellular Uptake of Entrapped Drug (Curcumin). Colloids Surf. A Physicochem. Eng. Asp. 2019, 567, 86–95. [Google Scholar] [CrossRef]
  282. Hassani, A.; Mahmood, S.; Enezei, H.H.; Hussain, S.A.; Hamad, H.A.; Aldoghachi, A.F.; Hagar, A.; Doolaanea, A.A.; Ibrahim, W.N. Formulation, Characterization and Biological Activity Screening of Sodium Alginate-Gum Arabic Nanoparticles Loaded with Curcumin. Molecules 2020, 25, 2244. [Google Scholar] [CrossRef]
  283. Nogueira, L.F.B.; Cruz, M.A.E.; Tovani, C.B.; Lopes, H.B.; Beloti, M.M.; Ciancaglini, P.; Bottini, M.; Ramos, A.P. Curcumin-Loaded Carrageenan Nanoparticles: Fabrication, Characterization, and Assessment of the Effects on Osteoblasts Mineralization. Colloids Surf. B Biointerfaces 2022, 217, 112622. [Google Scholar] [CrossRef]
  284. Xie, H.; Xiang, C.; Li, Y.; Wang, L.; Zhang, Y.; Song, Z.; Ma, X.; Lu, X.; Lei, Q.; Fang, W. Fabrication of Ovalbumin/κ-Carrageenan Complex Nanoparticles as a Novel Carrier for Curcumin Delivery. Food Hydrocoll. 2019, 89, 111–121. [Google Scholar] [CrossRef]
  285. Fan, Y.; Yi, J.; Zhang, Y.; Yokoyama, W. Fabrication of Curcumin-Loaded Bovine Serum Albumin (BSA)-Dextran Nanoparticles and the Cellular Antioxidant Activity. Food Chem. 2018, 239, 1210–1218. [Google Scholar] [CrossRef] [PubMed]
  286. Liu, Q.; Jing, Y.; Han, C.; Zhang, H.; Tian, Y. Encapsulation of Curcumin in Zein/ Caseinate/Sodium Alginate Nanoparticles with Improved Physicochemical and Controlled Release Properties. Food Hydrocoll. 2019, 93, 432–442. [Google Scholar] [CrossRef]
  287. Yao, K.; Chen, W.; Song, F.; McClements, D.J.; Hu, K. Tailoring Zein Nanoparticle Functionality Using Biopolymer Coatings: Impact on Curcumin Bioaccessibility and Antioxidant Capacity under Simulated Gastrointestinal Conditions. Food Hydrocoll. 2018, 79, 262–272. [Google Scholar] [CrossRef]
  288. Sorasitthiyanukarn, F.N.; Muangnoi, C.; Ratnatilaka Na Bhuket, P.; Rojsitthisak, P.; Rojsitthisak, P. Chitosan/Alginate Nanoparticles as a Promising Approach for Oral Delivery of Curcumin Diglutaric Acid for Cancer Treatment. Mater. Sci. Eng. C 2018, 93, 178–190. [Google Scholar] [CrossRef]
  289. Sorasitthiyanukarn, F.N.; Muangnoi, C.; Rojsitthisak, P.; Rojsitthisak, P. Chitosan-Alginate Nanoparticles as Effective Oral Carriers to Improve the Stability, Bioavailability, and Cytotoxicity of Curcumin Diethyl Disuccinate. Carbohydr. Polym. 2021, 256, 117426. [Google Scholar] [CrossRef]
  290. Wathoni, N.; Meylina, L.; Rusdin, A.; Mohammed, A.F.A.; Tirtamie, D.; Herdiana, Y.; Motoyama, K.; Panatarani, C.; Joni, I.M.; Lesmana, R.; et al. The Potential Cytotoxic Activity Enhancement of α-Mangostin in Chitosan-Kappa Carrageenan-Loaded Nanoparticle against MCF-7 Cell Line. Polymers 2021, 13, 1681. [Google Scholar] [CrossRef]
  291. Rathore, P.; Arora, I.; Rastogi, S.; Akhtar, M.; Singh, S.; Samim, M. Collagen Nanoparticle-Mediated Brain Silymarin Delivery: An Approach for Treating Cerebral Ischemia and Reperfusion-Induced Brain Injury. Front. Neurosci. 2020, 14, 538404. [Google Scholar] [CrossRef] [PubMed]
  292. Roy, S.; Rhim, J.-W. Fabrication of Chitosan-Based Functional Nanocomposite Films: Effect of Quercetin-Loaded Chitosan Nanoparticles. Food Hydrocoll. 2021, 121, 107065. [Google Scholar] [CrossRef]
  293. Li, H.; Wang, D.; Liu, C.; Zhu, J.; Fan, M.; Sun, X.; Wang, T.; Xu, Y.; Cao, Y. Fabrication of Stable Zein Nanoparticles Coated with Soluble Soybean Polysaccharide for Encapsulation of Quercetin. Food Hydrocoll. 2019, 87, 342–351. [Google Scholar] [CrossRef]
  294. Liang, X.; Cao, K.; Li, W.; Li, X.; McClements, D.J.; Hu, K. Tannic Acid-Fortified Zein-Pectin Nanoparticles: Stability, Properties, Antioxidant Activity, and in vitro Digestion. Food Res. Int. 2021, 145, 110425. [Google Scholar] [CrossRef]
  295. de Araujo, J.T.C.; Martin-Pastor, M.; Pérez, L.; Pinazo, A.; Sousa, F.F.O. de Development of Anacardic Acid-Loaded Zein Nanoparticles: Physical Chemical Characterization, Stability and Antimicrobial Improvement. J. Mol. Liq. 2021, 332, 115808. [Google Scholar] [CrossRef]
  296. Lima, R.A.; de Souza, S.L.X.; Lima, L.A.; Batista, A.L.X.; de Araújo, J.T.C.; Sousa, F.F.O.; Rolim, J.P.M.L.; Bandeira, T.D.J.P.G. Antimicrobial Effect of Anacardic Acid–Loaded Zein Nanoparticles Loaded on Streptococcus Mutans Biofilms. Braz. J. Microbiol. 2020, 51, 1623–1630. [Google Scholar] [CrossRef]
  297. Bernal-Mercado, A.T.; Juarez, J.; Valdez, M.A.; Ayala-Zavala, J.F.; Del-Toro-Sánchez, C.L.; Encinas-Basurto, D. Hydrophobic Chitosan Nanoparticles Loaded with Carvacrol against Pseudomonas Aeruginosa Biofilms. Molecules 2022, 27, 699. [Google Scholar] [CrossRef] [PubMed]
  298. Jain, A.; Sharma, G.; Kushwah, V.; Ghoshal, G.; Jain, A.; Singh, B.; Shivhare, U.S.; Jain, S.; Katare, O.P. Beta Carotene-Loaded Zein Nanoparticles to Improve the Biopharmaceutical Attributes and to Abolish the Toxicity of Methotrexate: A Preclinical Study for Breast Cancer. Artif. Cells Nanomed. Biotechnol. 2018, 46, 402–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  299. Abid, M.; Naveed, M.; Azeem, I.; Faisal, A.; Faizan Nazar, M.; Yameen, B. Colon Specific Enzyme Responsive Oligoester Crosslinked Dextran Nanoparticles for Controlled Release of 5-Fluorouracil. Int. J. Pharm. 2020, 586, 119605. [Google Scholar] [CrossRef] [PubMed]
  300. Adeel, M.; Duzagac, F.; Canzonieri, V.; Rizzolio, F. Self-Therapeutic Nanomaterials for Cancer Therapy: A Review. ACS Appl. Nano Mater. 2020, 3, 4962–4971. [Google Scholar] [CrossRef]
  301. Abnoos, M.; Mohseni, M.; Mousavi, S.A.J.; Ashtari, K.; Ilka, R.; Mehravi, B. Chitosan-Alginate Nano-Carrier for Transdermal Delivery of Pirfenidone in Idiopathic Pulmonary Fibrosis. Int. J. Biol. Macromol. 2018, 118, 1319–1325. [Google Scholar] [CrossRef] [PubMed]
  302. Thomas, D.; Mathew, N.; Nath, M.S. Starch Modified Alginate Nanoparticles for Drug Delivery Application. Int. J. Biol. Macromol. 2021, 173, 277–284. [Google Scholar] [CrossRef]
  303. Constantin, M.; Bucatariu, S.; Sacarescu, L.; Daraba, O.M.; Anghelache, M.; Fundueanu, G. Pullulan Derivative with Cationic and Hydrophobic Moieties as an Appropriate Macromolecule in the Synthesis of Nanoparticles for Drug Delivery. Int. J. Biol. Macromol. 2020, 164, 4487–4498. [Google Scholar] [CrossRef]
  304. Singh, T.A.; Sharma, A.; Tejwan, N.; Ghosh, N.; Das, J.; Sil, P.C. A State of the Art Review on the Synthesis, Antibacterial, Antioxidant, Antidiabetic and Tissue Regeneration Activities of Zinc Oxide Nanoparticles. Adv. Colloid Interface Sci. 2021, 295, 102495. [Google Scholar] [CrossRef]
  305. Horie, M.; Tabei, Y. Role of Oxidative Stress in Nanoparticles Toxicity. Free Radic. Res. 2021, 55, 331–342. [Google Scholar] [CrossRef]
  306. Scutera, S.; Argenziano, M.; Sparti, R.; Bessone, F.; Bianco, G.; Bastiancich, C.; Castagnoli, C.; Stella, M.; Musso, T.; Cavalli, R. Enhanced Antimicrobial and Antibiofilm Effect of New Colistin-Loaded Human Albumin Nanoparticles. Antibiotics 2021, 10, 57. [Google Scholar] [CrossRef]
  307. Lee, N.-Y.; Ko, W.-C.; Hsueh, P.-R. Nanoparticles in the Treatment of Infections Caused by Multidrug-Resistant Organisms. Front. Pharmacol. 2019, 10. [Google Scholar] [CrossRef]
  308. Benoit, D.S.W.; Sims, K.R.; Fraser, D. Perspective: Nanoparticles for Oral Biofilm Treatments. ACS Nano 2019, 13, 4869–4875. [Google Scholar] [CrossRef]
  309. Yeh, Y.-C.; Huang, T.-H.; Yang, S.-C.; Chen, C.-C.; Fang, J.-Y. Nano-Based Drug Delivery or Targeting to Eradicate Bacteria for Infection Mitigation: A Review of Recent Advances. Front. Chem. 2020, 8, 286. [Google Scholar] [CrossRef]
  310. Hill, M.; Twigg, M.; Sheridan, E.A.; Hardy, J.G.; Elborn, J.S.; Taggart, C.C.; Scott, C.J.; Migaud, M.E. Alginate/Chitosan Particle-Based Drug Delivery Systems for Pulmonary Applications. Pharmaceutics 2019, 11, 379. [Google Scholar] [CrossRef]
  311. Liu, J.; Xiao, J.; Li, F.; Shi, Y.; Li, D.; Huang, Q. Chitosan-Sodium Alginate Nanoparticle as a Delivery System for ε-Polylysine: Preparation, Characterization and Antimicrobial Activity. Food Control 2018, 91, 302–310. [Google Scholar] [CrossRef]
  312. Falciani, C.; Zevolini, F.; Brunetti, J.; Riolo, G.; Gracia, R.; Marradi, M.; Loinaz, I.; Ziemann, C.; Cossío, U.; Llop, J.; et al. Antimicrobial Peptide-Loaded Nanoparticles as Inhalation Therapy for Pseudomonas Aeruginosa Infections. Int. J. Nanomed. 2020, 15, 1117–1128. [Google Scholar] [CrossRef] [PubMed]
  313. Yu, H.; Ma, Z.; Meng, S.; Qiao, S.; Zeng, X.; Tong, Z.; Jeong, K.C. A Novel Nanohybrid Antimicrobial Based on Chitosan Nanoparticles and Antimicrobial Peptide Microcin J25 with Low Toxicity. Carbohydr. Polym. 2021, 253, 117309. [Google Scholar] [CrossRef] [PubMed]
  314. Rahimivand, M.; Tafvizi, F.; Noorbazargan, H. Synthesis and Characterization of Alginate Nanocarrier Encapsulating Artemisia Ciniformis Extract and Evaluation of the Cytotoxicity and Apoptosis Induction in AGS Cell Line. Int. J. Biol. Macromol. 2020, 158, 338–357. [Google Scholar] [CrossRef]
  315. Beconcini, D.; Fabiano, A.; Zambito, Y.; Berni, R.; Santoni, T.; Piras, A.M.; Di Stefano, R. Chitosan-Based Nanoparticles Containing Cherry Extract from Prunus Avium L. to Improve the Resistance of Endothelial Cells to Oxidative Stress. Nutrients 2018, 10, 1598. [Google Scholar] [CrossRef]
  316. Hadidi, M.; Pouramin, S.; Adinepour, F.; Haghani, S.; Jafari, S.M. Chitosan Nanoparticles Loaded with Clove Essential Oil: Characterization, Antioxidant and Antibacterial Activities. Carbohydr. Polym. 2020, 236, 116075. [Google Scholar] [CrossRef]
  317. Vahedikia, N.; Garavand, F.; Tajeddin, B.; Cacciotti, I.; Jafari, S.M.; Omidi, T.; Zahedi, Z. Biodegradable Zein Film Composites Reinforced with Chitosan Nanoparticles and Cinnamon Essential Oil: Physical, Mechanical, Structural and Antimicrobial Attributes. Colloids Surf. B: Biointerfaces 2019, 177, 25–32. [Google Scholar] [CrossRef]
  318. Song, X.; Wang, L.; Liu, T.; Liu, Y.; Wu, X.; Liu, L. Mandarin (Citrus reticulata L.) Essential Oil Incorporated into Chitosan Nanoparticles: Characterization, Anti-Biofilm Properties and Application in Pork Preservation. Int. J. Biol. Macromol. 2021, 185, 620–628. [Google Scholar] [CrossRef] [PubMed]
  319. Bagheri, R.; Ariaii, P.; Motamedzadegan, A. Characterization, Antioxidant and Antibacterial Activities of Chitosan Nanoparticles Loaded with Nettle Essential Oil. J. Food Meas. Charact. 2021, 15, 1395–1402. [Google Scholar] [CrossRef]
  320. Shetta, A.; Kegere, J.; Mamdouh, W. Comparative Study of Encapsulated Peppermint and Green Tea Essential Oils in Chitosan Nanoparticles: Encapsulation, Thermal Stability, in-Vitro Release, Antioxidant and Antibacterial Activities. Int. J. Biol. Macromol. 2019, 126, 731–742. [Google Scholar] [CrossRef]
  321. Karimirad, R.; Behnamian, M.; Dezhsetan, S. Application of Chitosan Nanoparticles Containing Cuminum Cyminum Oil as a Delivery System for Shelf Life Extension of Agaricus Bisporus. LWT 2019, 106, 218–228. [Google Scholar] [CrossRef]
  322. Yoncheva, K.; Benbassat, N.; Zaharieva, M.M.; Dimitrova, L.; Kroumov, A.; Spassova, I.; Kovacheva, D.; Najdenski, H.M. Improvement of the Antimicrobial Activity of Oregano Oil by Encapsulation in Chitosan—Alginate Nanoparticles. Molecules 2021, 26, 7017. [Google Scholar] [CrossRef]
  323. Cai, M.; Zhang, G.; Wang, J.; Li, C.; Cui, H.; Lin, L. Application of Glycyrrhiza Polysaccharide Nanofibers Loaded with Tea Tree Essential Oil/ Gliadin Nanoparticles in Meat Preservation. Food Biosci. 2021, 43, 101270. [Google Scholar] [CrossRef]
  324. Brotons-Canto, A.; González-Navarro, C.J.; Gil, A.G.; Asin-Prieto, E.; Saiz, M.J.; Llabrés, J.M. Zein Nanoparticles Improve the Oral Bioavailability of Curcumin in Wistar Rats. Pharmaceutics 2021, 13, 361. [Google Scholar] [CrossRef]
  325. Li, J.; Yang, Y.; Lu, L.; Ma, Q.; Zhang, J. Preparation, Characterization and Systemic Application of Self-Assembled Hydroxyethyl Starch Nanoparticles-Loaded Flavonoid Morin for Hyperuricemia Therapy. Int. J. Nanomed. 2018, 13, 2129–2141. [Google Scholar] [CrossRef]
  326. Das, S.; Das, M.K. Synthesis and Characterization of Thiolated Jackfruit Seed Starch as a Colonic Drug Delivery Carrier. Int. J. Appl. Pharm. 2019, 11, 53–62. [Google Scholar] [CrossRef]
  327. Larsson, S.; Jansson, M.; Boholm, Å. Expert Stakeholders’ Perception of Nanotechnology: Risk, Benefit, Knowledge, and Regulation. J. Nanopart. Res. 2019, 21, 57. [Google Scholar] [CrossRef]
  328. Yuan, H.; Guo, H.; Luan, X.; He, M.; Li, F.; Burnett, J.; Truchan, N.; Sun, D. Albumin Nanoparticle of Paclitaxel (Abraxane) Decreases While Taxol Increases Breast Cancer Stem Cells in Treatment of Triple Negative Breast Cancer. Mol. Pharm. 2020, 17, 2275–2286. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Classification of nanomaterials according to dimensionality (summarized from [2,6,13]).
Figure 1. Classification of nanomaterials according to dimensionality (summarized from [2,6,13]).
Ijms 24 03188 g001
Figure 2. Classification of nanomaterials based on their chemical composition (summarized from [7,16,18]).
Figure 2. Classification of nanomaterials based on their chemical composition (summarized from [7,16,18]).
Ijms 24 03188 g002
Figure 3. Biodistribution of the drug after intravenous administration when contained in biopolymeric nanoparticles (left) compared to the free drug (right) (summarized from [30]).
Figure 3. Biodistribution of the drug after intravenous administration when contained in biopolymeric nanoparticles (left) compared to the free drug (right) (summarized from [30]).
Ijms 24 03188 g003
Figure 4. Advantages and disadvantages of natural and synthetic polymers (summarized from [35,36,37]).
Figure 4. Advantages and disadvantages of natural and synthetic polymers (summarized from [35,36,37]).
Ijms 24 03188 g004
Figure 5. Classification of biopolymers based on their origin (summarized from [47,48]).
Figure 5. Classification of biopolymers based on their origin (summarized from [47,48]).
Ijms 24 03188 g005
Figure 6. Schematic representation of most commonly used methods for producing biodegradable nanoparticles from biopolymers.
Figure 6. Schematic representation of most commonly used methods for producing biodegradable nanoparticles from biopolymers.
Ijms 24 03188 g006
Table 1. Examples of proteins used for the production of protein-based nanoparticles.
Table 1. Examples of proteins used for the production of protein-based nanoparticles.
Origin of ProteinProtein SourceProteinRef.
AnimalsOvalbumin and bovine serum albuminAlbumin[71]
MilkCasein[72]
Muscles, tendons, skin, bonesCollagen[73]
Bones, skin, connective tissueGelatin[74]
Hair, nails, skin, feathers, wool, hooves, beaks, hornsKeratin[75]
Silkworm Bombyx moriSilk fibroin and sericin[76]
PlantsWheatGliadin[77]
Corn kernelZein[78]
Table 2. Examples of polysaccharides used for the production of polysaccharide-based nanoparticles.
Table 2. Examples of polysaccharides used for the production of polysaccharide-based nanoparticles.
Origin of PolysaccharidePolysaccharide SourcePolysaccharideRef.
AnimalShells of arthropods such as crabs, prawns, shrimps, and lobstersChitosan[88]
PlantLarix spp.Arabinogalactan[89]
Agricultural trashes: sugarcane bagasse, rice husksCellulose[37,90]
Plant sources: wood, cotton, jute, hemp, bamboo, coconut, wheat, rice, grass, agave, flax[37,90]
Cyamopsis tetragonolobaGuar gum[91]
Cyamopsis psoraloides[92]
Globe artichoke, topinambur, elecampane, chicoryInulin[93]
Citrus peels, apple pomacePectin[94]
Potatoes, corn, tubers, fruits, legumes, roots, cereals, rice, yams, banana wheat, sorghumStarch[37]
Plant tissues (trees, cotton)Cellulose[95]
AlgaeNizimuddinia zanardiniAlginate[96]
Sargassum latifolium[97]
Cystoseira barbata[98]
Ascophyllum nodosum[99]
Laminaria digitata[100]
Laminaria hyperborea[101]
Laminaria japonica[102]
Macrocystis pyrifera[103]
Sargassum wightii[104]
Chondrus crispusCarrageenan[105]
Sarcothalia crispata[106]
Gigartina skottsbergii[106]
Eucheuma denticulatum[107]
Kappaphycus alvarezii[108]
Hypnea musciformis[109]
FungiAureobasidium pullulansPullulan[110]
Cytaria harioti[111]
Cytaria darwinii[111]
Cryphonectria parasitica[111]
Termella mesenterica[112]
Teloschistes falvicans[112]
Rhodotorula bacarum[112]
Saccharomyces cerevisiaeDextran[113]
Rhizopus oryzaeChitosan[114]
BacteriaLactobacillus plantarumDextran[115]
Weissella confuse[116]
Weissella cibaria[117]
Leuconostoc mesenteroides[118]
Leuconostoc reuteri[117]
Lactobacillus gasseri[119]
Sphingomonas elodeaGellan gum[120]
Sphingomonas paucimobilis[120]
Leuconostoc citreumLevan[121]
Bacillus subtilis[122]
Bacillus atrophaeus[123]
Acinetobacter nectaris[124]
Xanthomonas campestrisXanthan gum[125]
Gluconacetobacter xylinusCellulose[126]
Azotobacter spp.Alginate[127]
Pseudomonas spp.[127]
Table 3. Examples of animal-based biopolymeric nanoparticles for healthcare applications.
Table 3. Examples of animal-based biopolymeric nanoparticles for healthcare applications.
BiopolymerPreparation MethodParticle Size (nm)Loaded CargoHealthcare ApplicationRef.
HSASelf-assembly method<200.0DocetaxelDelivery of anticancer drug[174]
HSAHigh-pressure homogenization150.4Prednisolone and curcuminDrug delivery for the treatment of rheumatoid arthritis[130]
HSADesolvation method<200.0PiperineDrug delivery for the treatment of breast cancer cells (MCF-7)[175]
Self-assembly method
HSACoacervation method85.0–120.0Imipenem and ciprofloxacinDelivery of antibiotics[176]
BSAEmulsification method100.0-Drug delivery studies[177]
BSADesolvation method156.0–315.0DoxorubicinDelivery of anticancer drug[178]
BSAAnti-solvent precipitation method150.0Paclitaxel and resveratrolDelivery of anticancer drug[134]
BSADesolvation method100.0–200.0Methylene blueDelivery of antifungal drugs for the treatment of Candida infections[135]
CollagenElectrospinning/electrospray method--Appropriate mimicry of the extracellular matrix for biomedical applications[142]
CollagenFilm dispersion method35.2–107.0Cucurbitacin BOral delivery[179]
ChitosanIonic gelation method106.5Zinc gluconateDrug delivery for the treatment of rheumatoid arthritis[167]
ChitosanEmulsion-ionic gelation40.0–100.0Clove essential oilSuperior inhibitory effect against Aspergillus niger[180]
ChitosanIonic gelation method75.4RotigotineNose-to-brain drug delivery[168]
ChitosanEmulsion-ionic gelation198.7–373.4Basil essential oilDelivery of antibacterial substances[181]
ChitosanIonic gelation method208.1CinnamaldehydeDelivery of anti-quorum sensing agents[182]
ChitosanIonic gelation method174.0–898.0Pomegranate peel extractControlled delivery of natural antioxidants and antimicrobials[183]
ChitosanEmulsification-ionic gelation method174.0–293.0Lemongrass essential oilDelivery of antibacterial substances[184]
Table 4. Examples of plant-based biopolymeric nanoparticles for healthcare applications.
Table 4. Examples of plant-based biopolymeric nanoparticles for healthcare applications.
BiopolymerPreparation MethodParticle Size (nm)Loaded CargoHealthcare ApplicationRef.
GliadinDesolvation method1.0–181.0SumatriptanBrain-targeted drug delivery[188]
GliadinElectrospray method481.6γ-OryzanolDrug delivery to reduce HT-29 cell proliferation[191]
ZeinNanoprecipitation method120.0–180.0ResveratrolOral drug delivery[195]
ZeinLiquid–liquid phase separation method379.4BMP6-derived peptideBone regenerative medicine[196]
ZeinPhase separation37.0–112.3MaytansineDelivery of anticancer drug[219]
ZeinSelf-assembly method180.0Thymbra capitata essential oilDrug delivery[220]
PectinIonotropic gelation method700.0–850.0-Potential oral drug delivery[200]
PectinIonic gelation method482.7MetforminDrug delivery for type 2 diabetes mellitus[221]
StarchNanoprecipitation method500.0QuercetinEstimation of release kinetics[204]
StarchNanoprecipitation method<250.0CurcuminDrug delivery for polyphenols[222]
StarchNanoprecipitation method255.0–396.0Ginkgo biloba extractsOral drug delivery[223]
StarchEmulsification method300.05-FluorouracilColon-specific drug delivery[224]
StarchAcid hydrolysis and precipitation method85.1PaclitaxelDelivery of anticancer drug[225]
StarchNanoprecipitation method105.0ParacetamolpH-responsive drug delivery system[205]
StarchMicroemulsification method282.9Triphala ChurnaDrug delivery of drugs with antimicrobial, antibiofilm, and neuroprotective effects[226]
CelluloseNanoprecipitation method70.0–365.0Methylene blueSustained and controlled release[227]
Guar gumEmulsification method200.0CelecoxibOral drug delivery[214]
InulinEmulsion solvent evaporation method224.0–365.0-Intracellular stimulation of probiotics[228]
InulinSpray drying method289.8QuercetinColon-targeted drug delivery[217]
Table 5. Examples of algal-based biopolymeric nanoparticles for healthcare applications.
Table 5. Examples of algal-based biopolymeric nanoparticles for healthcare applications.
BiopolymerPreparation MethodParticle Size (nm)Loaded CargoHealthcare ApplicationRef.
AlginateIonic gelation method-RifampicinDrug delivery without systemic toxicity[244]
AlginateEmulsification method279.1MiltefosineAntifungal drug delivery [236]
AlginateEmulsification method150.0–270.0Betamethasone sodium phosphateOcular drug delivery[245]
AlginateIonic gelation method400.0–1000.0Grape pomace extractOral delivery[246]
AlginateElectrospray method228.0CRISPR plasmids DNAGene delivery[247]
AlginateEmulsification method279.1Bacteriocins from L. paracaseiDrug delivery for the treatment of fungal diseases caused by Candida spp. and Cryptococcus spp.[236]
κ-CarrageenanElectrospray method309.5–663.8D-limoneneDrug delivery of highly sensitive bioactive agents[242]
κ-CarrageenanEmulsification method211.0CurcuminDrug delivery[248]
Table 6. Examples of fungal-based biopolymeric nanoparticles for healthcare applications.
Table 6. Examples of fungal-based biopolymeric nanoparticles for healthcare applications.
BiopolymerPreparation MethodParticle Size (nm)Loaded CargoHealthcare ApplicationRef.
PullulanNanoemulsification method98.0Ursodeoxycholic acidDrug delivery for neuroprotective effect[257]
PullulanSelf-assembly method86.4–222.3MitoxantroneDelivery of anticancer drug[253]
PullulanNanoprecipitation method<200.0ValsartanDelivery of cardiovascular agents[258]
PullulanEmulsion solvent evaporation method200.0DoxorubicinLiver-specific drug delivery[259]
PullulanSelf-assembly method185.7Methotrexate and 10-hydroxycamptothecineTumor-targeted drug delivery[260]
PullulanProbe-sonication method84.1ErlotinibDrug delivery for cervical cancer therapy[261]
Table 7. Examples of bacterial-based biopolymeric nanoparticles for healthcare applications.
Table 7. Examples of bacterial-based biopolymeric nanoparticles for healthcare applications.
BiopolymerPreparation MethodParticle Size (nm)Loaded CargoHealthcare ApplicationRef.
DextranSpray drying method-RifampicinEnhanced deep lung delivery[272]
DextranIonic gelation method34.0–42.0-Ocular drug delivery[265]
DextranSingle emulsion evaporation method99.8–181.35, 10, 15, 20-tetraphenyl-21H, 23H-porphyrineDrug delivery for a tumor cell-targeted photodynamic therapy[273]
DextranSelf-assembly method260.0NaproxenAnti-inflammatory drug delivery[274]
DextranSolvent diffusion method<70.0Chloroquine diphosphateDelivery of antimalarial drug[275]
DextranIonic gelation method69.3-Antibacterial agents[276]
Xanthan gumAnti-solvent precipitation149.0–241.0Cinnamon bark extractControlled delivery of polyphenols[277]
LevanElectrohydrodynamic atomization method82.1ResveratrolDrug delivery for wound healing and tissue engineering[269]
Gellan gumNebulization/ionic gelation method337.1ResveratrolOral drug delivery[268]
Gellan gumIonic gelation method85.6AtenolDelivery of antihypertensive drug[270]
Table 8. Examples of therapeutic agents loaded into biopolymeric nanoparticles for different biomedical purposes.
Table 8. Examples of therapeutic agents loaded into biopolymeric nanoparticles for different biomedical purposes.
Incorporated Therapeutic CompoundBiopolymerAdministration RouteTherapeutic ApplicationRef.
Bioactive compounds
SylmarinCollagenIntraperitoneal injectionEnhanced neuroprotective effect against acute ischemia/reperfusion injury[291]
Cucurbitacin BCollagenOral deliveryEnhanced absorption of orally administered Cucurbitacin B[179]
CurcuminBSA/dextranIn vitroEnhancement of cellular antioxidant activity of curcumin in Caco-2 cells[285]
CurcuminZeinOral administrationIncreased oral bioavailability[324]
CurcuminZeinIntravenous injectionPotential drug delivery platform for therapy of glioblastoma[193]
Beta-caroteneZeinOral administrationEnhanced cellular uptake, cytotoxicity, and improved oral biopharmaceutical performance[298]
Anacardic acidZeinIn vitroEnhanced antimicrobial activity[295]
QuercetinZein stabilized with soluble soybean polysaccharideIn vitroPotential all-natural delivery systems for bioactive molecules[293]
MorinStarchIntravenous injectionTherapeutic efficacy in the treatment of hyperuricemia[325]
Conventional drugs
Prednisolone and curcuminHSAIntravenous injectionImproved in vivo therapeutic effect in rats with rheumatoid arthritis[130]
Paclitaxel and resveratrolBSAIntravenous injectionImproved suppression of tumor growth without systemic toxicity[66]
Paclitaxel and di-fluorinated curcuminBSAIntravenous injectionA promising platform for treating gynecological cancers[133]
RotigotineChitosanNose-to-brain administrationEfficient drug delivery as an alternative to conventional routes[168]
Zinc gluconateChitosanIntraperitoneal injectionA potential alternative for the treatment of rheumatoid arthritis[167]
PirfenidoneChitosan/alginateTransdermal administrationDrug delivery for the treatment of idiopathic pulmonary fibrosis[301]
RifampicinAlginateOral administrationNo systemic toxicity and excellent safety after oral administration of nanoparticles[244]
MaytansineZeinIntravenous injectionStrong anti-A549 tumor cell activity[219]
SumatriptanGliadinNasal administrationEffective crossing of the blood-brain barrier[188]
MetforminPectinIn vitroOral administration for management of Type 2 Diabetes Mellitus[221]
IbuprofenStarchOral administrationColon-targeted drug delivery[326]
MitoxantronePullulanIn vitroInhibition of the growth of bladder cancer cells (MB49)[253]
Ursodeoxycholic AcidPullulanIn vitroIncreased neuroprotective effect[257]
PaclitaxelDextranIntravenous injectionPotential for C6 glioma treatment[266]
Chloroquine diphosphateDextranParenteral administrationTreatment of malaria[275]
Antibiotics and antimicrobial agents
ColistinHSAIn vitroHigh antibacterial activity against the growth and inhibition of biofilm formation of multidrug-resistant Gram-negative bacterial species[306]
TobramycinAlginate/chitosanIn vitroEfficient delivery of antimicrobial drug[310]
ε-PolylysineAlginate/chitosanIn vitroDelivery of antibacterial agents[311]
SET-M33 peptideDextranIntrapulmonary administrationEfficient treatment of lung infection[312]
Extracts
Pomegranate peel extractChitosanIn vitroEnhanced antioxidant and antimicrobial properties[183]
Grape pomace extractChitosan/alginateOral deliveryImproved biological activity[246]
Artemisia ciniformis extractAlginateIn vitroInhibition of proliferation of AGS cells[314]
Ginkgo biloba extractStarchIn vitroSustained release in artificial gastric and intestinal juices[223]
Essential oils
Oregano essential oilAlginate/chitosanDermal administrationEnhanced antimicrobial activity[322]
Clove essential oilChitosanIn vitroImproved antioxidant and antibacterial activity[316]
Basil essential oilChitosanIn vitroEnhanced antimicrobial activity and strong antibiofilm activity[181]
Mandarin essential oilChitosanIn vitroEnhanced inhibition of biofilm formation[318]
Lemongrass and peppermint essential oilChitosanIn vitroEnhanced thermal stability, antioxidant, and antimicrobial activity[184]
Green tea essential oilChitosanIn vitroEnhanced thermal stability, antioxidant, and antimicrobial activity[320]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kučuk, N.; Primožič, M.; Knez, Ž.; Leitgeb, M. Sustainable Biodegradable Biopolymer-Based Nanoparticles for Healthcare Applications. Int. J. Mol. Sci. 2023, 24, 3188. https://doi.org/10.3390/ijms24043188

AMA Style

Kučuk N, Primožič M, Knez Ž, Leitgeb M. Sustainable Biodegradable Biopolymer-Based Nanoparticles for Healthcare Applications. International Journal of Molecular Sciences. 2023; 24(4):3188. https://doi.org/10.3390/ijms24043188

Chicago/Turabian Style

Kučuk, Nika, Mateja Primožič, Željko Knez, and Maja Leitgeb. 2023. "Sustainable Biodegradable Biopolymer-Based Nanoparticles for Healthcare Applications" International Journal of Molecular Sciences 24, no. 4: 3188. https://doi.org/10.3390/ijms24043188

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