Next Article in Journal
Evolution Mechanism of Sputtered Film Uniformity with the Erosion Groove Size: Integrated Simulation and Experiment
Next Article in Special Issue
Chitosan Soft Matter Vesicles Loaded with Acetaminophen as Promising Systems for Modified Drug Release
Previous Article in Journal
Comparative Analysis of Volatile Organic Compound Purification Techniques in Complex Cooking Emissions: Adsorption, Photocatalysis and Combined Systems
Previous Article in Special Issue
Cytotoxicity Enhancement of α-Mangostin with Folate-Conjugated Chitosan Nanoparticles in MCF-7 Breast Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Strategies for the Preparation of Chitosan Derivatives for Antimicrobial, Drug Delivery, and Agricultural Applications: A Review

by
Rajeev Shrestha
1,*,
Anusree Thenissery
1,
Rahul Khupse
2 and
Gireesh Rajashekara
1,*
1
Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA
2
College of Pharmacy, University of Findlay, Findlay, OH 45840, USA
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(22), 7659; https://doi.org/10.3390/molecules28227659
Submission received: 28 September 2023 / Revised: 14 November 2023 / Accepted: 15 November 2023 / Published: 18 November 2023
(This article belongs to the Special Issue Chitosan, Chitosan Derivatives and Their Applications)

Abstract

:
Chitosan has received much attention for its role in designing and developing novel derivatives as well as its applications across a broad spectrum of biological and physiological activities, owing to its desirable characteristics such as being biodegradable, being a biopolymer, and its overall eco-friendliness. The main objective of this review is to explore the recent chemical modifications of chitosan that have been achieved through various synthetic methods. These chitosan derivatives are categorized based on their synthetic pathways or the presence of common functional groups, which include alkylated, acylated, Schiff base, quaternary ammonia, guanidine, and heterocyclic rings. We have also described the recent applications of chitosan and its derivatives, along with nanomaterials, their mechanisms, and prospective challenges, especially in areas such as antimicrobial activities, targeted drug delivery for various diseases, and plant agricultural domains. The accumulation of these recent findings has the potential to offer insight not only into innovative approaches for the preparation of chitosan derivatives but also into their diverse applications. These insights may spark novel ideas for drug development or drug carriers, particularly in the antimicrobial, medicinal, and plant agricultural fields.

1. Introduction

Over the last few decades, natural polymers, including biopolymers, polyesters, and proteins, have displayed diverse applications in the fields of agriculture, pharmaceuticals, food safety, and the cosmetic industry due to their biodegradability, eco-friendliness, compatibility, and cost-effectiveness [1,2,3]. As a result, natural polymers have gained significantly increased interest in creating novel drugs, owing to their distinct and intricate chemical composition, which is responsible for their wide and diverse range of biological effects [4,5]. Among these, chitosan (CS) stands out as a widely utilized polysaccharide that has been extensively studied across various scientific disciplines [6,7,8]. CS is composed of linear polysaccharides with varying amounts of (β1 → 4) linked residues of N-acetyl-2 amino-2-deoxy-D-glucose (glucosamine, GlcN) and 2-amino-2-deoxy-D-glucose (N-acetyl-glucosamine, GlcNAc) residues distributed inside the polymer. It is derived from chitin, polyβ-[1,4]-N-acetyl-d-glucosamine) (or poly(N-acetyl-d-glucosamine) via deacetylation in the presence of sodium hydroxide or enzymes (Scheme 1) [9,10,11]. Chitin is found in crustaceans, including crabs, shrimp, shellfish, and lobsters, and its extraction primarily involves demineralization, deproteinization, and discoloration [12,13]. Its hydrophilic and reactive properties are limited by the presence of acetyl groups (CH3CO) attached to the amino groups of GlcNAc. These acetyl groups function not only as barriers for water molecules, but also as protective shields, which reduce the availability of the amino groups for reactions [14]. For the biological and physical activity of CS, the degree of deacetylation (DD) is the pivotal factor, which represents the proportion of β-1,4-D-glucosamine repeating units in the polysaccharides during the deacetylation process originating from chitin [9,10,11,14]. A higher DD denotes an increased presence of free amino groups, which can form ammonium (NH4+) groups within a pH range of 5–6, resulting in increased water solubility and the enhancement of the biological impact of CS [15]. Furthermore, a lower molecular weight (MW) is another factor that decreases with a higher DD, which impacts the bioactivity of CS [15]. CS with a lower MW has demonstrated antibacterial, antioxidant, and anti-tumor properties due to its increased water solubility, ease of permeability to cells, and binding efficacy to DNA or RNA [16,17].
CS is a cationic biopolymer agent that can interact with negatively charged substances such as lipids, fats, ions, cholesterol, proteins, and other organic and inorganic molecules [18]. CS was first identified in 1811 in mushrooms. Extensive research has revealed that CS significantly exhibits antimicrobial, antiviral, anticancer, non-toxic, water treatment, biocompatible, and biodegradable properties [19,20]. However, its solubility in acidic conditions (pH less than 6) limits certain bioactivities and applications. To address these issues and to enhance the bioactive and physiochemical effectiveness under neutral and physiological conditions, the modification of CS is a promising alternative since it has three active functional groups: C2-NH2, C3-OH, and C6-OH (Scheme 1) [21]. The nucleophilic substitution reactions involving the amino and hydroxyl groups, followed by multi-step reactions, have been studied, including N-alkylation/amination, acetylation, cross-linking, oxidation, grafting, and sulfation [22,23]. Synthetic reactions are accomplished through chemical or enzymatic catalysis, polymerization, or hydrolysis conditions to achieve the desired product [24]. The CS derivatives, which vary in terms of their degree of substitution (DS), a crucial factor in the structure–activity relationship (SAR), demonstrate unique physiochemical characteristics. Novel scaffolds with these characteristics have enhanced biocompatibility, bioactivity, biodegradability, and non-toxicity, while retaining the inherent pharmacological effects of their antibacterial, anticancer, and antiviral properties [25]. Moreover, CS derivatives have also been widely utilized in the field of nanomaterials due to their immense potential as carriers for targeted drugs, adjuvants, or vaccines, facilitating the treatment of various diseases including cancer, diabetes, Alzheimer’s, cardiovascular disorders, and inflammatory conditions [26,27]. These nanomaterials have shown superior outcomes compared to conventional drug formulations.
Herein, we explore the recent progress of CS derivatives, focusing on modifications to their functional groups conjugated to the polymer skeleton as well as substitutions in the active sites, including C2-NH2, C3-OH, and C6-OH. Importantly, we discuss the synthesis process of CS derivatives, categorizing them into various chemical groups based on their synthetic pathway or functional characteristics, which included alkylated, acylated, Schiff base, quaternary ammonia, guanidine, and heterocyclic CS derivatives. Moreover, this review also comprehensively covers the diverse applications of CS and its derivatives as nanomaterials in the antimicrobial, agricultural, and drug delivery fields, including the elucidation of their mechanisms of action and the factors that contribute to superior outcomes compared to conventional approaches.

2. Global Research Trends of Chitosan

The global research trends surrounding CS underscore its significant scientific and technological value. As projected by Precedence Research, the CS market is expected to surpass US $29 billion by 2030, reflecting its substantial importance across various sectors [28]. These trends are driven by several key factors. Notably, there is a growing demand for CS in various end-user industries, an increasing emphasis on global water treatment activities, significant advancements in healthcare and the medical sector in industrialized countries, and a heightened awareness of public health concerns, including rising obesity rates. The Asia–Pacific region has assumed a prominent position, dominating the CS market share [28]. Over the last two decades, CS has ascended to a leading role in research involving biomedicine, nanomedicine, and cell and tissue engineering [29]. The increase in research endeavors is well noted by the heightened number of patents, particularly in the biomedical sector, where its gelling properties enable viscosity and density adjustments under specific physiochemical conditions to enhance biocompatibility [30] (Table 1). Furthermore, the applications of CS underscore these global research trends. CS finds utility in diverse fields, which have extended constant research efforts in drug delivery, dentistry, ophthalmology, wound dressings (some marketed CS-based wound dressings: HemCon® Bandage PRO, HemCon ChitoFlex® PRO, Chitopack C, ChiGel, Tegaderm, Tegasorb, and Traumastat), cell encapsulation, bioimaging, tissue engineering, food packaging, gelling and coating, abiotic stress prevention in plants, enhanced water availability in crops, controlled-release fertilizers, dye-sensitized solar cells, wastewater and sludge treatment, and metal extraction [31,32]. This multifaceted use highlights CS’s pivotal role in shaping global research trends across healthcare, agriculture, and various industries on a worldwide scale, making it a central focus in the realm of biopolymers.

3. Preparation Methods of Chitosan Derivatives

Owing to the efficient biological and physiochemical properties of CS derivatives, various pathways of CS modification have been explored. The methods involve single- to multi-step reactions, including direct modification, catalytic or enzymatic reactions, chemical grafting, and cross-linking reactions under various conditions [23]. In terms of the chemical reactivity of CS, 2-NH2 displays more reactivity compared to the OH group. Therefore, to achieve the selective functionalization of the OH group, NH protection by using the phthaloyl group has been employed [48,49,50]. However, OH protection groups, including triphenylmethyl [51], trimethylsilyl [52], and tertiary-butyl dimethylsilyl [53,54], have also been utilized to achieve the specific functionalization of the NH group, resulting in a high DS in the products.
Although CS is soluble in acidic water through the binding process of H+ with the N atom of amino group, CS exhibits a high degree of crystallinity due to the intermolecular and intramolecular hydrogen bonding, leading to its near insolubility in normal water [55]. Therefore, the key benefit of CS modification is the enhancement of the water solubility of CS in high pH ranges by introducing hydrophilic groups like amino, carbonyl, carboxyl, hydroxyl, or sulfhydryl groups into the CS [55,56,57,58,59]. These functional groups are capable of not only destroying the original hydrogen bonding and crystallinity of CS but also creating new hydrogen bonds with water, thereby increasing the solubility of the product. Consequently, the modified CS is often employed to improve the solubility of the product, demonstrating the synergetic efficacy of attached functional groups for antimicrobial, antifungal, and other biological activities [23,24]. The researchers have employed CS with a wide range of MWs (ranging from 50 to 1400 kDa) and DDs (ranging from 70 to 95%) in their experiments to produce CS derivatives with various ranges of DSs of the functional group. A higher DS leads to better water solubility and higher biological potentials [29,60].

3.1. Alkylation, Acylation, and Schiff Base-Based Chitosan Derivatives

The alkylation of CS entails the hydrophobic modification of this material. The resulting hydrophobic traits yield specific benefits, especially the restriction of liquid interchange between the external surroundings and wound tissue. This property creates a protective barrier on the wound surface that impedes the infiltration of airborne bacteria and upholds gas exchange, ultimately leading to rapid wound healing [61]. The alkylation of CS (2a) is conducted by employing various alkyl or allyl halides such as bromoethane, bromohexane, bromododecane, bromohexadecane, chlorobutane, and allyl bromide in the presence of a strong base like sodium hydroxide (Scheme 2) [62,63,64]. However, the Nie group reported that the Michael addition reaction of CS with hydroxyethylacryl in acidic media led to alkylated CS (2b) [65]. On the other hand, CS has been acylated with various acyl chlorides in the presence of NaOH, with the initial step involving the dissolution of CS in acetic acid with water (2c) [66,67]. The acylation reaction has the potential to disrupt both the intramolecular and intermolecular H-bonding within the CS, leading to a reduction in its crystallinity and enhancing water solubility [68]. In recent studies, researchers have explored the reaction of CS with pre-prepared acyl chlorides or acetic acid containing diverse heterocycles like benzothiazole, thiadiazole, thymine, or pyrazoles to produce the acylated CS bearing the heterocycles (2d, 2e) [69,70,71,72,73,74,75,76]. Similarly, Zhang’s group developed CS-bearing quinolinyl urea derivatives (2f). The reaction was accomplished by initiating a reaction between methyl chloroformate and CS, followed by a subsequent reaction with 2-aminoquinoline [77]. Specifically, the incorporation of heterocyclic groups into CS, with substitution degrees of 57.4% [75] and 47% [76], resulted in enhanced antimicrobial activities.
In the course of the modification of CS derivatives by using CS (MW: 100–300 kDa, DD: 74–95%), the preparation of Schiff bases emerges as the most straightforward method. This approach includes the reaction of an amine group with benzaldehydes through an intermediate carbinolamine (Scheme 3). This method not only facilitates the introduction of new groups, such as heterocycles, but also enables subsequent alkylation after reduction [78,79]. In a plethora of studies, the attachment of heterocycles such as thiophene, furan, pyrazole, indoles, quinolone, benzothiazole, oxazole, and imidazole-bearing Schiff base CS derivatives (2g; DS: 1.15–56%) was extensively explored in acetic acid as a mild condition [80,81,82,83,84]. Incorporating heteroatoms into CS as Schiff base resulted in a hydrophobic characteristic; nevertheless, the presence of pyrazole and thiophene groups exhibited water solubility for CS [80,81,82,83,84]. Additionally, the reduction of Schiff bases with sodium borohydride has been widely employed as an alternative alkylation strategy, leading to the attachment of new functional groups or heterocycles (DS: 36–81%) (2h) [85]. Moreover, Khrustalev et al. revealed innovative CS derivatives with novel heterocyclic structures (2i), forming complexes with platinum metals [86]. Herein, CS of various MWs (viscosity average MW: 3.7 × 104 D to 17.8 × 104 D) was employed to introduce nitrile derivatives of CS (2i’) in the range of DS, 17% to 59%. This synthesized platinum complex was not soluble in water; however, the 1,2,4-oxadiazoline CS derivatives prepared from nitrones, a functional group consisting of N-oxide and imine, were soluble in water and showed a high antibacterial activity coupled with low toxicity [86].

3.2. Quaternary Ammonium Chitosan Derivatives

Quaternary ammonium CS derivatives are the modified form of CS salt bearing a positively charged nitrogen atom, covalently attached with alkyl or aryl groups. The hydrophilic quaternary ammonium group enhances the water solubility of CS by introducing a quaternary ammonium salt group. The salt increases the electrostatic charge and weakens intramolecular hydrogen bonds, resulting in a high level of water solubility [87]. This quaternary ammonium salt improves not only the hydrophilicity of the final CS derivatives but also enhances its antimicrobial properties, making it valuable in diverse applications, including wound dressings, drug carriers, and other biomedical fields [88]. In addition, the polymeric quaternary ammonium CS bearing heterocyclic compounds has attracted a significant antimicrobial agent due to the synergetic effects of CS, quaternary ammonia salts, and heterocyclic compounds [89]. Commonly, the quaternary ammonium salt, N-trimethyl CS (TMC, 3a), is prepared via the methylation of CS (MW: 5–200 kDa; DD: 73–97%) in the presence of sodium iodide and iodomethane under alkaline conditions (Scheme 4) [90,91,92,93,94,95,96,97,98,99]. However, there is the possibility of O-methylation, which can be reduced by using either an acidic or heterogeneous medium. The inclusion of heterocyclic and aromatic moieties shows promising prospects for drug development [91]. By taking advantage of this, TMC has been further subjected to derivatization, enabling the incorporation of heterocyclic and aromatic groups. In this respect, Guo et al. prepared the intermediate 6-bromo-6-deoxy-N-trimethyl quaternary ammonium CS through the bromination of N-bromobutanimide (NBS) on TMC. Subsequently, it was further reacted with coumarins with electron-donating or electron-withdrawing groups under basic conditions to yield coumarins bearing TMC (DS: 69–82%) (3b) [92]. In another study of their group, 6-bromo-6-deoxy-N-trimethyl quaternary ammonium CS was further investigated by treating it with ethylenediamine and mono- or bi-substituted polyhydroxyl benzaldehyde in two steps, leading to the formation of phenol or polyphenol-containing trimethyl quaternary ammonium CS (3c) [93].
In the meantime, an alternative approach has been introduced to prepare O-chloroacetyl quaternary ammonium CS, which has been further employed in chemical modifications involving heterocyclic compounds due to its high effectiveness in nucleophilic substitution reactions (Scheme 4). For example, compounds such as quinoline (DS: 24.34–98.7%), imidazole (DS: 12.7–65.5%), amino pyridines (DS: 72–82%), pyridines, substituted pyridines bearing Schiff bases (DS: 21.5–58.3%) or urea derivatives (DS: 21–91%), and tertiary amines (DS: 45.3–84.3%) were subjected to reactions with O-chloroacetyl quaternary ammonium CS, resulting in the formation of double quaternary ammonium CS salt derivatives (3d3h) [96,97,98,99]. The advantages of double-quaternary ammonium salt include making CS more cationic and highly soluble in water.
While developing the novel derivatives of quaternary ammonium CS, the water-soluble (3-Chloro-2-hydroxypropyl) trimethy/triethyl ammonium chloride (CHTAC) or glycidyl trimethyl ammonium chloride serves as an excellent quaternary ammonium source, which has been employed in nucleophilic substitution reactions, either C2-NH2 or C6-OH, of CS (Scheme 5) [100,101,102,103,104,105,106,107,108]. In recent years, n-alkyl quaternary ammonium CS (3i) and 2-hydroxypropyl trimethyl ammonium chloride CS (3j’) were synthesized through the reaction of n-alkyl CS or CS with a quaternary ammonium source, CHTAC. The n-alkyl CS was initially obtained through the alkylation of CS with n-butylaldehyde using the Schiff alkali method, followed by reduction using sodium borohydride [100]. The optimal synthesis condition for a higher DS (70–90%) of 3i was optimized in various temperatures, molar ratios of chemicals, and reaction times. While the extended n-alkyl chain notably reduces the intermolecular hydrogen bonds in CS, it alone does not lead to complete dissolution in water. However, when combined with quaternary ammonium salt, it significantly enhances water solubility [100]. On the other hand, 3j’ was further dialyzed with carboxymethyl insulin [101] and other acetate derivatives against deionized water [102], followed by lyophilization to obtain the desired quaternary ammonium CS derivatives (3j; DS: 17.5–40.9% and 46–81%). In the meantime, the Zheng group also explored imidazole-based quaternary ammonium CS derivatives (DS: 36%) through multi-step nucleophilic substitution reactions [103]. The synthesis process comprised a sequential generation of intermediary compounds, N-chlorobutanol CS and N-(1-carboxybutyl-4-(3-methyl-imidazole) CS chloride, through 4-chlorobutanol chloride and N-methylimidazole. The ultimate product (3k) was obtained through the reaction between CHTAC and N-(1-carboxymethyl-4-(3-methyl-imidazole)) CS chloride. Likewise, hydroxypropyl trimethyl ammonium chloride CS (HACC) has demonstrated itself as an excellent water-soluble quaternary CS derivative, which is commonly prepared from the reaction of CS and glycidyl trimethyl ammonium chloride in deionized water [104,105]. Furthermore, the introduction of thioctate [106] and Schiff base [107] functionalities onto HACC was accomplished through treatments with α-lipoic acid and 4-diethylaminobenzaldehyde to give the new derivatives 3l (DS: 51–86%) and 3m. In an alternate approach to the aforementioned procedure, the researchers initially established a Schiff base of CS, followed by a reaction with glycidyl trimethyl ammonium chloride, yielding O-quaternary ammonium N-benzylidene CS. To expand the scope of chemical modifications, O-quaternary ammonium N-acyl thiourea CS (3n; DS: 81–93%) was synthesized through the incorporation of ammonium thiocyanate and chloroacetyl chloride into the reaction sequence [108].
Another category of quaternary ammonium salt is the pyridinium salt group, which acts as a cationic surfactant, is soluble in water [87,109], and exhibits noteworthy antimicrobial and antifungal activities [110,111]. Pyridine functions as a nucleophile, capable of replacing the alkyl halide to synthesize the quaternary pyridinium compound [112]. Typically, CS undergoes modification through a reaction with either the halide group or the pyridine group, enabling subsequent functionalization with pyridine salts (Scheme 6) [113]. Huang et al. grafted pyridine to N-chlorobutyryl CS, resulting in the formation of pyridinium CS salt (3o, DS: 38%) [114]. N-chlorobutyryl CS was prepared from the reaction of CS and 4-chlorobutyryl chloride via acylation. Recently, Tan et al. synthesized CS-bearing pyridinium salt along with quaternary ammonium salt (DS: 35–97%) (3p) through a treatment between CS and narcotic acid, followed by a reaction with bromopropyl trialkyl ammonium bromides [115]. In another study, Guo et al. also introduced CS derivatives that contain pyridine-bearing Schiff bases (3q, DS: 22.7–50%) [116] and heterocyclic scaffolds along with thiourea (3r, 3s; DS: 21–91%, 18–88%) [98,117]. In contrast with these conventional approaches, the Zincke reaction has been explored for the direct transformation of primary amines into N-aryl pyridinium salt [118]. This process typically involves the use of N-(2,4-dinitrophenyl)-pyridinium chloride, commonly known as Zincke salt, as the reagent, which follows an SN(ANRORC) mechanism involving nucleophilic substitution addition, ring opening, and subsequent ring closure [119]. In this respect, the CS amino group at the C2 position was directly converted to CS pyridinium salt (3t) via the Zincke reaction [120,121].
The application of click chemistry has demonstrated significant utility in introducing quaternary ammonium salts into CS molecules, primarily due to its notable benefits of high efficiency and selectivity [122]. For instance, the initial step involved the development of N-trimethyl ammonium CS, which also served as a C2-NH2 protection group. Then, the reaction was followed by either azidation [123] or O-acetylation [124,125] to proceed with the cycloaddition reaction of alkyne-azides, resulting in the integration of water soluble triazole substituents with quaternary ammonium CS (2u, 2v; DS: 72–83%, 28–92%, 53–92%), which exhibited antifungal and antioxidant properties (Scheme 7).

3.3. Guanidine Chitosan Derivatives

Guanidines have demonstrated a significant interest as a valuable functional group incorporated with multiple drugs for various therapeutic applications and biological activities, such as antimicrobial, antifungal, antimalarial, and others [126]. From a chemical perspective, guanidines function as the nitrogenous analog of carbonic acid, behaving as strong organic bases with higher pKa values. However, by introducing the appropriate substituents, such as CS, onto the nitrogen atom, the basicity of guanidine can be adjusted, and it is highly soluble in neutral aqueous solutions by disrupting hydrogen bonding and hydrophobic effects in polymers, resulting in the remarkable versatility of guanidine motif-bearing scaffolds [55,127]. Water-soluble CS-bearing guanidines (4a, 4b; DS: 8.4–14.2%, 14–30%) are commonly synthesized through the reaction of cyanimide or aminoiminomethanesulfonic acid with CS in the presence of hydrochloric acid (Scheme 8) [128,129]. In the alternative pathway, the microwave-assisted grafting of guanidine oligomers with CS was also investigated, and the thus-formed CS guanidine (4c) in various DSs was utilized for the production of hygiene paper products [130]. Moreover, the main composition of the peptide, L-arginine, or L-arginine Schiff base, was also coupled with CS in a basic medium to develop biomimetic CS derivatives (4d; DS: 6–61.5%) for the improvement of the permeable capabilities of cell-penetrating peptides, wound healing, and antimicrobial and antioxidant activities [131,132,133,134,135].
Furthermore, CS–guanidine derivatives incorporating additional moieties such as poly (3-hydroxy butyrate) (4e) or quaternized carboxymethyl (4f; DS: 16–73%) were also synthesized (Scheme 8) [136,137]. In this process, cyanoguanidine or cyanimide functioned as the source of the guanidine. However, the reactions involved multiple sequential steps of the nucleophilic reaction of poly(3-hydroxybutyrate) or epichlorohydrin. Another approach for synthesizing guanidinylated CS derivatives involved the utilization of protective groups like tertiarybutyl dimethylsilyl (TBDMS) and tertiarybutyloxycarbonyl (Boc). These protective groups effectively overcome the limitation of selective modification of the amino group with a high degree of substitution. In the course of the reaction, the TBDMS group was initially introduced to the hydroxyl group of CS possessing a free amino group, enabling the successful synthesis of a 100% guanidinium substitution of the amino group, thereby yielding the desired guanidinium CS derivatives (4g, 4h; DS: 13–100%) [138]. In the exploration of diverse derivatives, the Mingshan Li group functionalized the CS nanofiber membrane with the polyhexamethylene guanidine group (4i). The synthesis process included the oxidation of CS in the presence of sodium periodate (NaIO4), electrospinning with polyvinyl alcohol (PVA), and functionalization with polyhexamethylene guanidine [139]. Thus, the formed nanofiber membrane exhibited not only antibacterial activities but also high adsorption capacities of Cu (II) and Congo red.
In contrast to the aforementioned method, Peter Hesemann et al. employed a different approach to produce an N-guanidinium CS derivative. This was carried out in the presence of scandium (III) triflate and acetic acid, followed by a subsequent sol–gel reaction utilizing 3-(trihydroxysilyl)-1-propanesulphonic acid to synthesize a guanidinium CS silica hybrid material (4j) (Scheme 9) [140]. This material demonstrated remarkable effectiveness as an adsorbent for removing dyes from wastewater. On the other hand, the same group investigated the reaction between CS and carbodiimides in ionic liquid media in the presence of 1-butyl-3-methylimidazolium chloride (BMIM Cl) to produce N-guanidinium CS (N,N′-dicyclohexyl) chloride (4k) and N-guanidinium CS (N-(3-dimethylaminopropyl)-N′-ethyl hydrochloride) chloride (4l), showing significant amicrobial properties [141]. Similarly, the Sedghi group devised a semi-conductive water-soluble biguanidine/polyaniline composite (4m; DS: 18.6%) through the reaction of CS with cyanoguanidine followed by aniline incorporation (Scheme 10). Subsequently, the composite was further introduced into a self-healing waterborne polyurethane system, with shape-memory effect properties suitable for bone tissue engineering [142].

3.4. Heterocyclic Chitosan Derivatives via Click Chemistry or N-Functionalized Reaction

Copper-mediated click chemistry has emerged as a versatile approach for synthesizing triazole moieties that involve the cycloaddition reaction between an azide and terminal alkyne, allowing for the incorporation of diverse bioactive functional groups into biomacromolecule backbones [143,144]. Furthermore, this strategy serves as a stable antimicrobial pharmacophore, displaying its dual significance [145]. In this regard, CS has been widely functionalized with heterocyclic scaffolds, such as substituted triazole groups formed through click reactions, and demonstrated significant antimicrobial and antifungal activities [49,124,146,147,148,149,150,151]. The primary advantage of incorporating triazole, a polar moiety, is its excellent water solubility, which imparts hydrophilic properties to the resulting CS derivatives bearing triazole groups [146,147,148,149,150,151]. CS has been modified by introducing an azido group or alkyne group into the 2-N or 6-O position for the click reaction (Scheme 11). The Li group introduced the azido group at both active positions of CS through the reaction of chloroacetyl chloride, followed by sodium azide, and subsequently reacted with prop-2-yn-1-yl nicotinate in the presence of copper sulfate to achieve antifungal (1,2,3-triazol-4-yl)methyl nicotinate CS (5a) [151]. The phthalolyl group was utilized for the protection of the 2-NH2 group, facilitated the 6-O position for azidation through stepwise bromination and azidation reactions, and was followed by the 3+2 cycloaddition reaction to provide diverse CS derivatives with triazoles (5b) [49,152]. Alternatively, another approach involved azidation through a nucleophilic NH2 reaction with compounds like 1-azido-3-chloro-2-propanol or 2(azidomethyl) oxiran.
Subsequent reactions with substituted acetylene in the presence of a Cu catalyst also led to the triazole-functionalized CS (5c) [153,154]. In addition, alkylation on the 2-N position through CS mesylate salt [155] or direct N-azidation in the presence of sodium nitrite and sodium azide [156,157] also enabled the click reactions to synthesize CS triazoles (5d, 5e, 5f).
In a further study of CS-based heterocycles, the researchers developed cross-linked CS-based hydrogels by using Diels–Alder reactions (Scheme 12). Furan-modified CS, which was obtained via the reaction of furfural with CS, was then subjected to the Diels–Alder click reaction with poly(ethylene)glycol-maleimide derivatives or pre-synthesized maleimide-functionalized CS in acidic conditions to prepare the hydrogels (5g, 5h; DS: 21–31%) with the capability of controlled drug release [158,159]. Moreover, the researchers have also constructed CS azalactone gel (5i) or genepin-crosslinked CS hydrogel (5j) through the direct substitution of oxygen with the nitrogen of the NH2 group of CS (Scheme 13) [160,161]. In another approach for attaching the heterocycles to CS, the direct reaction of isobenzofurandione or pyrano-quinolinedione derivatives with CS led to the CS-heterocycles 5k5n (DS: 7–18%) and 5o (DS: 34.79–92.05%) through either the direct interaction with the NH2 group or the ring opening of benzofurandiones [162,163].

4. Applications of Chitosan Derivatives

4.1. Antimicrobial Actions of Chitosan

CS has garnered extensive attention across a wide spectrum of applications, ranging from its role as an antimicrobial agent to its utility as a drug carrier. Numerous research investigations have sought to unveil CS’s antimicrobial attributes against a diverse array of microorganisms, including bacteria, fungi, yeast, and algae, primarily due to its biocompatible, biodegradable, and non-toxic properties [20,164]. The antimicrobial effects of CS are rooted in its physicochemical characteristics, which encompass the degree of deacetylation, its structural advantage in possessing reactive hydroxyl groups at the C-3 and C-6 positions, environmental conditions, and the specific microorganism in question [164]. The native CS, chitin, exhibits limited antimicrobial efficacy due to the presence of acetylated repeating units, which restrain the exposure of free amino groups. Furthermore, it is highly hydrophobic in nature because of the strong intra- and intermolecular hydrogen bonding [165]. On the other hand, CS-bearing repeating units of β-(1 → 4)-2-amino-2-deoxy-β-D-glucose offer primary amine functionality, typically achieved through the deacetylation process. A higher DD in the CS during the deacetylation process signifies an increased presence of free amino groups. These amino groups can interact with microbial cell surfaces through electrostatic attraction. Additionally, CS’s solubility in acidic water allows it to form cationic polyelectrolytes, facilitating its easy interaction with microbial cells and leading to cell lysis and cell death [166]. High-molecular-weight and low-molecular-weight CS exhibit distinct actions against microbes. High-molecular-weight CS’s antimicrobial activity primarily resides in the extracellular compartment, as it cannot penetrate the cell wall. Here, it operates by altering cell permeability, resulting in a reduced nutrient uptake from the extracellular milieu and a diminished availability of essential metals due to chelation. In contrast, low-molecular-weight CS displays the capability to exert both extracellular and intracellular effects. This translates to alterations in mitochondrial function and consequential impacts on RNA and protein synthesis [167,168].
CS has undergone extensive testing against both Gram-positive and Gram-negative bacteria for antibacterial activity (Figure 1, Table 2). However, the precise mechanism of action remains incompletely understood. Most studies underscore CS’s polycationic nature as a key contributor to its antibacterial efficacy [169]. Furthermore, its interactions with the bacterial cell wall are also dependent on the pH of the environment. When the environmental pH falls below CS’s pKa, electrostatic interactions occur between the polycationic CS and the anionic components of the bacterial cell wall. These components include lipopolysaccharides in the Gram-negative cell wall and certain proteins on the cell surface of bacteria. These interactions lead to the exertion of antibacterial effects [22,169]. Interestingly, the extent of electrostatic interactions is largely influenced by the number of amino groups linked to C-2 on the CS backbone. A higher number of amino groups results in more positively charged sites, enhancing the interaction with the carboxyl-negative charges on the bacterial cell wall [170].
Conversely, when the environmental pH exceeds the pKa, electrostatic interactions weaken, and chelating and hydrophobic effects come into play for antibacterial activity [169,170]. When chelation effects outweigh electrostatic forces, metal ions like Fe2+, Cu2+, and Zn2+ present on the bacterial surface can be chelated by CS’s amino groups [164,169,171]. Many of these divalent cations play a vital role in stabilizing bacterial cell membranes through their interactions with phosphate groups in teichoic acids and lipopolysaccharides (LPSs) in Gram-positive and Gram-negative bacteria, respectively [172,173]. Variations in susceptibility to CS exist between Gram-positive and Gram-negative bacteria due to marked differences in their cell wall compositions. Gram-positive bacterial cell walls consist of peptidoglycan, teichoic acids, and lipoteichoic acids [174,175]. These components contribute to high-density negative charges within the cell wall, hindering ion movement across the membrane. Positively charged CS can interact with these negatively charged teichoic acids in peptidoglycans, leading to cell membrane damage and the release of intracellular constituents [22,176]. Studies have also demonstrated that interactions with CS can result in peptidoglycan hydrolysis, intensifying electrostatic interactions, and enhancing antibacterial activity [177].
In Gram-negative bacteria, CS targets negatively charged lipopolysaccharides, causing the disruption of the outer membrane, the penetration of the cell membrane, and ultimately, bacterial death [178,179]. High-molecular-weight CS can form a dense polymer film on the bacterial cell surface, obstructing porins and impeding nutrient exchange, which ultimately leads to bacterial cell death [178,180]. Evidence of this is observed in scanning electron microscope studies, which revealed vesicle-like structures on the outer membrane of CS-treated Escherichia coli (E. coli) and Salmonella typhimurium (S. typhimurium) [181]. On the contrary, low-molecular-weight CS can penetrate bacterial cells, affecting DNA, RNA, and protein synthesis [20]. Studies have shown that when E. coli has been exposed to oleoyl-CS nanoparticles, the nanoparticles bind to DNA and RNA and influence the electrophoretic mobility of both nucleic acids [171]. CS and its derivatives have been extensively explored for their antimicrobial properties against various pathogens, offering diverse mechanisms of action [182]. Thioether CS oligosaccharides and CS–silver nanocomposites have been explored for their synergistic antimicrobial effects against multiple Gram-positive and Gram-negative bacteria [183,184]. Furthermore, another study reported the increased antibacterial activity of N-(2-ethylamino)-CS and N-2(2,6-diaminohexanamide)-CS polymers, attributed to the presence of amino groups and their hydrophobic interactions [185]. Similar studies have introduced CS Schiff bases with aromatic substitutions, which exhibit enhanced interactions with bacterial cell components [186]. CS Schiff bases were developed to potentially interact with specific components in both Gram-positive and Gram-negative bacteria, exerting antibacterial action [187]. The synthesis and antimicrobial properties of N-guanidinium CS derivatives were also investigated, highlighting their permeability-enhancing effects through adsorption and intracellular binding [141].
CS nanoparticles, when loaded with antibiotics, peptides, and proteins, have displayed promising results in possessing antibacterial effects and disrupting biofilm formation by bacteria. For instance, ceftriaxone sodium encapsulated within CS nanoparticles has exhibited heightened intracellular antibacterial efficacy against S. typhimurium [188]. Cefazolin-loaded CS nanoparticles have demonstrated enhanced antibacterial activity, particularly against multi-drug-resistant Gram-negative bacteria [189]. Meanwhile, tetracycline-loaded O-carboxymethyl CS nanoparticles have shown the potential to augment the action of antibiotics against Staphylococcus Aureus (S. aureus) infections [190]. Similarly, CS-based systems have been widely used for assessing their potential as carriers for probiotics or beneficial bacteria that can be used for controlling pathogenic bacterial infections. In one such study, our group demonstrated that CS–alginate nanoparticles carrying E. coli Nissle effectively inhibited Campylobacter jejuni growth in vitro [191].
Within the realm of CS metal nanocomposites, silver-based variants have received substantial attention. Several studies have revealed their effectiveness against a spectrum of bacteria, including S. aureus, E. coli, and S. typhimurium [190,192,193]. Furthermore, CS-based systems have been evaluated for their impact on biofilms. Anti-biofilm properties have been explored using preparations such as CS–streptomycin conjugates and CS–gold nanoparticles against various Gram-positive and Gram-negative bacteria. These studies have revealed that CS conjugation enhances the drug’s ability to disrupt biofilms by improving its penetration and contact with the bacterial surface [194]. Due to these antimicrobial properties, CS-based systems have undergone extensive investigation for their potential applications in wound healing, drug delivery carriers, and various biomedical applications.
The utilization of CS as an antimicrobial agent presents a dynamic landscape of potential research avenues and practical applications. Nanoparticle engineering is poised to harness CS’s inherent properties for the development of finely tuned nanoparticles, enabling precise, targeted antibacterial delivery with a focus on enhanced efficacy. Synergy exploration within combination therapies involving CS and other antimicrobial agents unveils an exciting frontier in the fight against antibiotic-resistant pathogens. In-depth investigations into the intricate molecular interactions governing CS’s biofilm disruption capabilities hold significant promise for novel strategies for combating persistent biofilm-associated infections. Concurrently, rigorous assessments of CS’s biocompatibility and toxicity profiles are vital, ensuring the safe integration of CS-based therapies into clinical practice. The vast potential of CS derivatives including CS oligomers and quaternized CS beckons, as they offer tailored solutions to specific antibacterial challenges. Elucidating the precise mechanisms underpinning CS’s antibacterial action, encompassing cell membrane interactions and resistance mechanisms, as well as understanding the host immune responses to CS, not only advances fundamental knowledge but also guides the rational design of more potent therapies in the future.
Table 2. Application of chitosan and its derivative for antibacterial activities.
Table 2. Application of chitosan and its derivative for antibacterial activities.
CS
Derivative/Preparations
BacteriaAssayEffect/Mechanism of ActionReferences
CS with benzoimidazolyl-thiadiazoleS. aureus, B. subtilis,
E. coli,
P. aeruginosa,
C. albicans
Agar well diffusion methodPresence of polar groups, sulfur, and nitrogen, increases the solubility and electrostatic attraction between polymer and bacterial cell wall and increases cell death[70]
Thiadiazole CS derivativeE. coli, P. aeruginosa,
B. subtilis, S. aureus
Agar well diffusion methodIncrease solubility due to cationic groups contributing to improved antibacterial activity[71]
1,3,4-thiadiazole modified CSE. coli, P. aeruginosa, B. subtilis, S. aureusAgar well diffusion methodIncrease solubility due to hydrophilic thiadiazole derivatives [72]
Thymine-modified CSP. aeruginosa,
A. baumannii, S. aureus, E. coli, MRSA
96-well plate microdilution method, scanning electron microscopyIncrease ζ-potential of the positively charged thiamine-modified CS derivatives; higher positive electric density; enhance electrostatic interaction with the negatively charged bacterial membrane[74]
CS linked with diphenyl pyrazole with succinic
anhydride
B. subtilis, S. aureus, P. aeruginosaAgar well diffusion methodPolycationic nature of CS and presence of amino group in pyrazole; penetration of CS into nucleus, blocking binding sites of RNA to DNA, inhibiting synthesis of cell wall proteins[75]
CS coupled with 4-((5, 5-dimethyl-3-oxocyclohex-1-en-1-yl) amino) benzene-sulfonamide (CS Schiff base)E. coli, S. aureusVisible spectroscopy analysis, MIC assayIncrease hydrophobicity of CS Schiff base, improving interaction with peptidoglycan[78]
CS with heteroaryl pyrazole derivatives (CS Schiff base)E. coli, K. pneumonia, S. aureus, S. mutansAgar well diffusion methodFunctional modification with pyrazole ring bearing pyridyl moiety enhanced antibacterial effect[80]
CS with formyl pyrazole derivatives (CS/pyrazole Schiff base)S. aureus, B. cereus,
E. coli
Agar well diffusion methodPresence of furan ring and nitro groups; enhance entry of CS to nucleus and interaction with RNA/DNA[81]
CS with indole-3-carboxaldehyde and 4-dimethylaminobenzaldehyde (phenolic CS Schiff bases)S. aureus, B. cereus,
E. coli, P. aeruginosa,
S. spp.
Agar well diffusion methodInteraction with bacterial cell membrane and disruption of cell wall integrity[82]
Chitooligosaccharide-niacin acid conjugateS. aureus, E. coli,
V. harveyi
Broth dilution assayIncrease lipophilicity, hydrophobic interaction of the trialkyl chain, increased interaction of quaternary ammonium salts and lipid structure of bacterial cell membrane[115]
Pyridine-4-aldehyde Schiff bases grafted chloracetyl CS oligosaccharide derivativesS. aureus, E. coliAgar well diffusion methodIncrease positive charge of the derivative and interaction with bacterial cell membrane[116]
L-arginine Schiff bases acylated CS derivativesB. cinerea, S. aureus, E. coliPlate colony counting methodFree positive charge and guanidine carried by CS combines with the negatively charged components of the bacterial cell wall[135]
N-guanidinium CS acetate, N-guanidinium CS chlorideE. coli, P. aeruginosa, S. aureus, B. subtilisTurbidimetric methodCationic groups enhance the permeability via adsorption followed binding to intracellular constituents[141]
Chitotriazolan (poly(β(1-4)-2-(1H-1,2,3-triazol-1-yl)-2-deoxy-D-glucose))S. aureus, E. coli96-well plate microdilution methodLoss of integrity to bacterial cell wall[146]
Calcium–CS–triazole nanocomplex E. coli, B. subtilisAgar well diffusion methodEnhance antibacterial activity contributed by triazole moiety[157]
CS hydrogel containing hydroxypropyl methylcellulose (HPMC)S. aureus,
P. aeruginosa
Biofilm assay/confocal scanning laser microscopyIncrease adhesiveness and penetration into biofilm and disruption[182]
Thioether CS oligosaccharide (CS oligosaccharide coupled with 3-bromopropene and tiopronin)S. aureus, B. subtilis,
L. monocytogenes,
E. coli, P. aeruginosa
MIC and MBC determination, cytotoxicity evaluation, antioxidant activity assaysEnhance antimicrobial capability due to carboxyl groups possessing positive charges, which bind to negatively charged cell membranes. Increased iron chelation from tiopronin.[183]
Biogenic CS–silver nanocompositeS. aureus, P. aeruginosa, S. spp., E. spp., S. spp., Shigella spp.Well diffusion technique for antimicrobial susceptibilitySynergistic effect of CS and silver; greater adsorption onto the surface of bacterial cells and can easily penetrate the bacterial cell wall causing cell death[184]
N-(2-ethylamino)-CS and N-2(2,6-diaminohexanamide)-CS polymersS. aureusBacterial growth
inhibition,
cytocompatibility studies
Presence of the amino groups: hydrophobic interaction with the bacterial wall is responsible for the enhanced activity[185]
CS Schiff base (CS with 2,4,6-trimethoxybenzaldehyde)S. aureus, E. coli,
P. aeruginosa,
K. pneumonia,
S. haemolyticus
Turbidity assayEnhance interaction of CS with peptidoglycan and plasma membrane due to aromatic substitution[186]
CS coupled with cyclohexanone and 2-N-methyl pyrrolidone (CS Schiff base)S. aureus, E. coli,
P. aeruginosa,
B. cereus
Agar well diffusion assay, MIC determination, bactericidal studiesInteraction with teichoic and lipoteichoic acid in Gram-positive bacteria and with O-specific side chain of LPS of Gram-negative bacteria[187]
CS with immobilized ZnO nanoparticlesE. coli, S. aureusAgar well diffusion methodAntibacterial activities[195]
Low-molecular-weight CSE. coli DH5α,
P. putida F1, L. lactis IO-1, B. subtilis 168
Zeta potential measurement, contact angle measurementAlteration of cell surface charge, cell surface hydrophobicity[196]
CS nanoparticlesP. spp.Disk diffusion test, antibiofilm assayCompromise structural integrity of the biofilms, heterogeneous destruction of the biofilm matrix[197]
Thiazolium CS (Quaternary CS)L. innocua,
S. epidermidis,
S. aureus, MRSA
Broth dilution methodCombine antibacterial effect of thiazolium and CS[198]
CS covalently bounded to isocyanate terminated quaternary ammonium salt and sulfopropylbetaine E. coli, S.aureusReal-time turbidity monitoring via automated turbidimetric systemNH2 group of the derivative binds with Mg2+ and Ca2+ in bacterial outer membrane[199]
Note: S. aureus: Staphylococcus aureus, B. subtilis: Bacillus subtilis, E. coli: Escherichia coli, P. aeruginosa: Pseudomonas aeruginosa, C. albicans: Candida albicans, A. baumannii: Acinetobacter baumannii, MRSA: Methicillin-resistant Staphylococcus aureus, K. pneumonia: Klebsiella pneumonia, S. mutans: Streptococcus mutans, B. cereus: Bacillus cereus, S. spp.: Salmonella spp., V. harveyi: Vibrio harveyi, B. cinerea: Botrytis cinerea, L. monocytogenes: Listeria monocytogenes, E. spp.: Enterococcus spp., S. haemolyticus: Staphylococcus haemolyticus, P. putida F1: Pseudomonas putida F1, E. coli DH5α: Escherichia coli DH5α, L. lactis IO-1: Lactococcus lactis IO-1, B. subtilis 168: Bacillus subtilis 168, L. innocua: Listeria innocua, S. epidermidis: Staphylococcus epidermidis.

4.2. Chitosan Derivatives as Drug or Vaccine Delivery System

Controlled drug or vaccine release is highly emphasized as a strategy for controlling disease pathologies, achieved through the targeted accumulation of therapeutic compounds at the site of the ailment or the utilization of inventive therapeutic agents to control infectious pathogens [200,201]. Drug encapsulation and dosage reduction emerge as the most convenient methods for achieving controlled drug release [202]. CS and its derivative-based nanoparticles and films have received considerable attention as highly suitable candidates for drug or antigen carriers due to their mucoadhesive properties, pH sensitivity, and biocompatibility. The release of drugs/antigens from CS nanoparticles in biological fluids occurs through diffusion, erosion, or swelling mechanisms [203] (Figure 2). Additionally, their distinct antimicrobial activities collaboratively synergize with drugs to enhance their antimicrobial effectiveness [204].
The recent applications of CS-based nanomaterials as drug carriers against infectious diseases are summarized in Table 3 [205,206,207,208,209,210,211,212,213,214,215,216,217,218]. For example, the nasal delivery of hesperidin-loaded CS nanoparticles exhibited enhanced cellular uptake within inflammatory microenvironments and effectively suppressed cytokine storm syndrome and acute lung injury [205]. Meanwhile, CS biguanidine nanoparticles displayed remarkable inhibition against various Mycobacterium tuberculosis strains, including sensitive, multidrug-resistant (MDR), and extensively drug-resistant (XDR) types, surpassing the performance of the standard anti-tuberculosis drug, isoniazid [206]. Sawant et al. demonstrated that tuberculosis prevention could be achieved through the pulmonary delivery of rifampicin with improved solubility when encapsulated within octanoyl CS nanoparticles [207]. Additionally, CS-based materials proved effective in delivering various anti-infectious drugs and antibiotics, including carvacrol [208], doripenem [209], chlorhexidine [210], ciprofloxacin [211], dolutegravir [212], endolysin Cpl-1 [213], and amphotericin B [214]. These materials successfully inhibited the growth of pathogens such as Salmonella enterica, Pseudomonas aeruginosa, Staphylococcus aureus, Staphylococcus epidermidis, Escherichia coli, human immunodeficiency virus HIV, and fungal species. In the expanded application, the eradication of waterborne pathogens such as rotavirus and Shigella spp. was also investigated through CS-wrapped carbon nanotubes [215].
In highlighting the effort in the encapsulation process, the inactivated virus, RNA, proteins, and probiotics have also been capsulated in the CS derivatives, which serve as a nano-vaccine delivery system to increase the immune response against the pathogens [219,220,221,222,223,224,225]. As an instance, Tafaghodi et al. encapsulated the inactivated PR8 influenza virus within CS alginate and trimethyl CS nanoparticles which exhibited immunoadjuvant capabilities after nasal immunization, thereby stimulating a Th1-type immune response [219]. Inactivated dengue virus (DENV)-loaded CS nanoparticles displayed their high immunogenicity, inducing elevated levels of antibodies and T cells against DENV-2 in comparison with soluble DENV-2 immunogens [220]. CS-based nanoparticles, containing Salmonella Enteritidis outer membrane proteins and flagellin proteins, emerged as a mucosal vaccine, inducing specific immune responses and reducing cecal colonization in avian hosts [221]. Our group also demonstrated the microencapsulation of probiotic E. coli Nissle 1917 within the CS–alginate, which reduced Campylobacter jejuni colonization in infected chickens [222].
Due to their cationic nature and high endocytosis potential, CS and its derivatives are not only utilized for drug and vaccine delivery for infectious diseases but also find applications in addressing various other conditions, such as cancer, gastrointestinal disorders, pulmonary ailments, Alzheimer’s disease, and diabetes [226,227,228,229,230,231,232,233,234,235,236,237,238,239]. Table 4 provides a summary of drug delivery systems designed to treat a range of diseases utilizing various forms of CS and its derivatives. CS-based nanoparticles enhance the drug-targeting ability and improve the blood retention time through desorption, swelling, and erosion. For instance, a PEGylated CS nanoparticle was employed to deliver a chemotherapeutic agent such as doxorubicin (DOX) in combination with breast cancer-specific monoclonal antibodies to treat breast cancer [226]. Thiolated CS nanoparticles delivered vincristine by binding to leaky prostate tumor cells, stimulating the receptors of these cells, and facilitating their internalization through endocytosis [227]. In another approach to the drug delivery system, the formulation of CS-based films and patches was employed in the ocular and transdermal delivery of the drugs brimonidine tartrate (BT) and tizanidine, respectively [231,232]. The CS-BT film significantly improved the transport of BT across the cornea, offering a potential alternative eye drop in the future, whereas the transdermal delivery of tizanidine facilitated efficient drug permeation through the skin, maintaining a therapeutic concentration. Moreover, CS and its derivatives have been fabricated through the crosslinking and polymerization methods to prepare hydrogels that have demonstrated a controlled, painless, and biocompatible transdermal release of therapeutic ingredients [233]. In an effort to find suitable carriers, CS-based micelles and liposomes have emerged as innovative tools to address the challenges associated with drug administration, especially the issues of low water solubility and limited drug permeability [234,235,236,237,238]. For example, the amphiphilic colloidal structures, especially CS-based micelles, including the CS-bearing thiourea group, incorporated with gold nanorods and carboxymethyl CS with a vitamin E succinate nano-micellar system (O-CMCTS-VES), effectively facilitated the release of drugs like paclitaxel and DOX for antitumor activities [234,235]. In addition, CS derivatives were employed to encapsulate liposomes, which possess minimal toxicity and cell-like membranes, along with drugs like resveratrol, aptamer-functionalized 5-fluorouracil, photosensitizer HPPH, and hypoxia-activated prodrug TH-302, enhancing the efficient targeted delivery of these drugs [236,237,238].
The drug-release characteristics of CS nanoparticles exhibit intricate responsiveness to pH alterations, rendering them a versatile tool for precision drug delivery. Research indicates that alterations in pH levels induce significant shifts in CS nanoparticle size and drug molecule release. In a study incorporating 5-fluorouracil (5-FU) within CS nanoparticles, it was observed that as the pH transitions from 3 to 5, there is a substantial increase in particle size, resulting in a continuous and rapid release of 5-FU. Conversely, when the pH exceeds 5, a notable reduction in particle size is evident, particularly at pH 7.4. This pH-responsive behavior can be attributed to the protonation of primary amino groups within the CS chain, leading to heightened electric density and increased repulsion forces between interconnected CS chains. Consequently, the surface density of protonated amino groups and the degree of protonization are adaptably responsive to shifts in solution pH [240]. CS nanoparticles undergo a reversible process of swelling and contraction, contingent upon the environmental pH, resulting in particle sizes fluctuating from approximately 450 nm to 150 nm [241]. The pH-sensitive surface charge reversal also facilitates enhanced cellular uptake and endolysosomal escape, extending blood circulation time, mitigating side effects, and optimizing drug delivery efficiency [242]. Such adaptability holds significant promise for targeted drug delivery, aligning with the varying pH levels found within the human body, which include gastric juice with a low pH of 1–1.5, the physiological environment exhibiting a pH of 7.4, the tumor extracellular microenvironment typically with a pH 6.5, and the pH within endosomes and lysosomes ranging from 5.0–6.2 to 4.0–5.0, respectively [242,243]. Moreover, studies highlight the enhancement of responsive properties through the integration of pH-sensitive molecules with CS, further streamlining drug delivery in response to environmental cues [244]. Additionally, investigations demonstrate that under acidic conditions (pH 3.5), there is a swift release of model drugs, while at slightly alkaline conditions (pH 7.4), the release rate is attenuated [245].
Despite the encouraging implications of CS and its derivatives as a drug or vaccine carrier system, they suffer from specific limitations. These limitations involve factors such as a restricted capacity to carry larger-sized drugs or vaccines, issues related to stability, the occurrence of burst drug release upon administration, limitations in targeting specific diseases, and the potential for an immune response in certain individuals. Nevertheless, ongoing research on the development of novel drug carriers that are more biodegradable, non-invasive, and have a high drug-loading capacity is actively employed to overcome these constraints. In the future, there is potential for designing CS derivative nanoparticles in a computational way that may not only be capable of their targeting efficacy but also have the ability to interact with proteins and signaling pathways. This system could potentially reduce the drug dosage requirements with minimal side effects and improve the patient’s condition.

4.3. Chitosan Derivatives in Plant Agriculture

CS and its derivatives have gained significant interest in the agriculture field, especially for their role in enhancing plant growth, stimulating root and shoot development, increasing crop yield, and supporting environmentally friendly farming practices (Figure 3) [246]. The cationic property of CS derivatives enables effective interaction with the negatively charged components of the bacterial cell membrane, resulting in bactericidal or bacteriostatic effects [247]. On the other hand, the chelating characteristic of CS also establishes it as a tremendous antifungal agent [248]. Similarly, it has also demonstrated its role as an elicitor by activating the defensive genes in plant disease control [249].
Considering the encouraging advantages, CS derivatives have been utilized in a wide range of crops, including vegetables, flowers, fruits, cereals, and medicinal plants, in various forms such as seed treatment, foliar spraying, coating fruits or vegetables, and delivering nutrients through nanomaterials [250] (Table 5). For example, Udayashankar group investigated the CS exogenous seed priming of cucumbers seeds, resulting in the inducement of phytohormone production and an increment of resistance against cucumber powdery mildew disease [251]. Recent studies demonstrated the use of CS through seed priming, seed soaking, or seedling application in mung bean [252], Platycodon grandiflorus [253], and Carum copticum L. [254] for reducing abiotic stress and increasing photosynthesis and antioxidant activity, resulting in efficient plant growth. Similarly, plant diseases such as root rot caused by Fusarium solani in fenugreek were also managed with seed treatment [255]. CS derivatives such as CS–ferulic acid, CS–caffeic acid conjugate, and carboxymethyl CS were explored for seed treatment or seed coating in cucumbers [256] or Prunus davidiana [257]. As a result, they either increased the antioxidant activity or enhanced the biomass, photosynthetic capacity, and absorption of phosphorus and potassium to improve plant growth. In further applications, CS nanoparticles have been significantly introduced in seed treatment [258]. For instance, Jogaiach et al. explored CS-derived nanoparticles in tomato seed treatment, which led to a remarkable increase in immune responses by accumulating defense enzymes and lignin to eradicate bacterial wilt disease [259]. CS incorporated with organic compounds or metals, such as garlic essential oil or silver-loaded nanoparticles, demonstrated its application in seed treatment on barley or chilies, which reduced the antifungal activities in plant growth [260,261].
CS and its derivatives, along with nanoparticles, were also employed as foliar sprays to reduce plant diseases, functioning as elicitors, plant protectors, or biostimulants [262,263]. For example, the foliar spraying of CS or its nanoparticles for tomatoes elicited an increment in shoot biomass and flower numbers, with reduced interaction with arbuscular mycorrhizal fungi (AMF) [264]. CS foliar spraying significantly reduced drought stress in bermudagrass by enhancing the turf quality, chlorophyll content, and leaf relative water content, and decreasing the level of electrolyte leakage, malonaldehyde, and hydrogen peroxide content [265]. In another study, the spray of CS derivatives such as CS lactate on Ocimum basilicum L. and Melissa officinalis L. accumulated the valuable phenolic group rosmarinic acid (RA) and increased the shoot biomass [266]. This showed the capability of herbal plants to produce functional foods. In addition, by using the CS–selenium nanoparticle, Skalicky et al. explored foliar spray with bitter melons, alleviating salt stress by enhancing antioxidant enzyme activity, elevating proline concentration, maintaining relative water content and K+, and reducing malonaldehyde and hydrogen peroxide levels, as well as sodium accumulation in plant tissues [267].
Despite the plethora of studies of CS applications demonstrating remarkable outcomes in enhancing plant growth, disease resistance, and stress tolerance, there always remains a requirement to determine the optimal techniques, dosage, and timing for its most effective use [246,268]. Through ongoing research, technological innovation, and eco-friendly techniques, CS has the capability to revolutionize harmless agricultural methods, which could pave the way for a more sustainable agricultural future.
Table 5. Chitosan and its derivatives for applications in plant agriculture.
Table 5. Chitosan and its derivatives for applications in plant agriculture.
CS or Its
Derivatives
FormPlantInterferenceReferences
CSSeed primingCucumber
  • Upregulation of phytohormones;
  • Resistance against cucumber powdery mildew disease.
[251]
CSSeed primingMung bean
  • Alleviate water stress, increase antioxidant system, improve growth.
[252]
CSSeed soakingP. grandiflorus
  • Enhances plant growth, photosynthesis, resistance, yield, and quality.
[253]
CSSeedling and callus cultureC. copticum L.
  • Improve plant tolerance to oxidative stress under salinity;
  • Elevation of essential oils and antioxidant activity.
[254]
CSSeed treatmentFenugreek
  • Controlling the root rot disease by reducing F. solani;
  • Increase seed germination.
[255]
CS–
ferulic acid and
CS–
caffeic acid conjugate
Seed treatmentCucumber
  • Increase plant growth;
  • Antioxidant activity.
[256]
Carboxymethyl CSSeed coatingP. davidiana
  • Increase the biomass and photosynthetic capacity;
  • Promote the absorption of phosphorus and potassium;
  • Decrease nitrogen uptake.
[257]
CS
nanoparticle
Seed treatmentTomato
  • Enhancing the immune response by accumulating the defense enzyme and lignin.
[259]
Garlic essential oil-loaded CS
nanoparticle
Seedling Barley
  • Antifungal activity;
  • Alternative to tebuconazole as seed-dressing agent.
[260]
CS silver nanocompositesSeed treatmentChili
  • Antifungal activity against anthracnose pathogen.
[261]
CS and its
nanoparticles
Foliar sprayTomato
  • Enhance plant growth and flowering.
[264]
CSFoliar sprayBermudagrass
  • Improve drought tolerance.
[265]
CS
lactate
Foliar sprayO. basilicum L. and
M. officinalis L.
  • Enhance the accumulation of valuable phytochemicals.
[266]
CS–selenium
nanoparticle
Foliar sprayBitter melon
  • Increased salinity tolerance in bitter melon plants.
[267]
Note: P. grandifloras: Platycodon grandifloras, C. copticum L.: Carum copticum L., P. davidiana: Prunus davidiana, O. basilicum L.: Ocimum basilicum L., M. officinalis L.: Melissa officinalis L.

5. Conclusions and Future Outlooks

CS is considered a promising polysaccharide renowned for its extensive array of potential applications in the fields of biology, medicine, water treatment, the food industry, cosmetics, and biodegradable packaging. Certain constraints persist, such as a lower degree of deacetylation and elevated molecular weight, which directly result in poor water solubility in neutral aqueous solutions. The modification of CS through different strategic pathways is one perspective that not only improves the water solubility but also enhances the bioactivity of the final product. However, the CS-bearing long chain or hydrophobic scaffolds may diminish the water solubility; nevertheless, they can still be effectively incorporated into films, fibers, or polymeric materials to enhance the hydrophobicity and stability of the materials [269]. This review highlighted the recent methodologies for synthesizing CS derivatives and their notable uses in antimicrobial functions, nanomaterials employed in drug delivery systems, and applications within the agricultural sector. Recent progress in the development of CS derivatives has been achieved through various methods, such as forming Schiff bases with heterocyclic compounds, generating quaternary ammonium salts and guanidines, employing click chemistry, and using them in direct N-functionalization reactions. There are also critical challenges, including separation, purity, yield, and stability, to developing new CS-bearing compounds.
Exploring CS’s use as an antimicrobial agent unmasks a dynamic array of research possibilities, especially in the medical and agricultural fields. CS demonstrates significant bioactivity in combating both animal and plant pathogens. The research on nanoparticle engineering involving CS and its derivatives as drug carriers has significantly demonstrated the effective delivery of drugs or vaccines to target diseases in the pharmaceutical field. Antimicrobial dressings based on CS and its derivatives have received FDA approval [270,271]. However, new applications as drug carriers are still awaiting approval due to insufficient research on their mutagenicity and genotoxicity. Simple and mild preparation of CS-based nanoparticles is always encouraged for ideal drug delivery systems due to their water solubility. In addition, researchers have observed that treating seeds or foliar sprays by applying CS derivatives or nanomaterials demonstrated effective results in enhancing plant growth, increasing crop yields, and advancing food security. This approach not only reduces the need for pesticides and fertilizers but also contributes to environmental pollution reduction and promotes the sustainability of entire agricultural systems. Nevertheless, the detailed molecular mechanisms by which they influence seeds and entire plants remain unclear, underscoring the need for further research into their genomic, proteomic, and metabolic effects.
Looking ahead, the development of novel CS derivatives through the incorporation of biologically active heterocyclic groups using recent techniques and methodologies involving different reaction mechanisms like CO, CN, or CH activation reactions is a highly promising prospect. However, research into the structure–activity relationship is an important factor in developing bioactive derivatives, along with a high degree of substitution of the functional groups. Researchers are consistently encouraged to explore novel reactions in conjugation with the structure–activity relationship. The exact mechanism of modified CS derivatives against antimicrobial activities has not been thoroughly explored; nevertheless, these effects are often generalized with unmodified CS. Advanced research regarding the mechanism of antimicrobial activities due to the specific functional group of CS derivatives is needed. Continuous research and development efforts are needed to enhance the water solubility, mitigate toxicity, and bolster the immune responses and functioning mechanisms of CS-based derivatives for applications in human drug delivery and agrochemicals. Moreover, contemporary technologies like computational and artificial intelligence can be used to serve not only in the design and synthesis of innovative CS derivatives and nanoparticles, but also as a means to approach the most realistic mechanisms for antimicrobial effects, drug delivery systems, and plant growth enhancement.

Author Contributions

G.R., R.S. and R.K. were responsible for the conception of this review work. R.S. and A.T. conducted the literature review, manuscript writing, and created the image. G.R., R.S., A.T. and R.K. reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The research in the Rajashekara laboratory was supported by the U.S. Department of Agriculture (USDA) National Institute for Food and Agriculture (NIFA) (grants number 2015-68004-23131 and 2020-6701-31401), and by the United States Department of Agriculture Specialty Crops Research Initiative project awards 2019-51181-30010 and 2022-51181-38242.

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. Paradowska-Stolarz, A.; Wieckiewicz, M.; Owczarek, A.; Wezgowiec, J. Natural Polymers for the Maintenance of Oral Health: Review of Recent Advances and Perspectives. Int. J. Mol. Sci. 2021, 22, 10337. [Google Scholar] [CrossRef]
  2. Gupta, R.K.; Guha, P.; Srivastav, P.P. Natural Polymers in Bio-Degradable/Edible Film: A Review on Environmental Concerns, Cold Plasma Technology and Nanotechnology Application on Food Packaging—A Recent Trends. Food Chem. Adv. 2022, 1, 100135. [Google Scholar] [CrossRef]
  3. Puertas-Bartolomé, M.; Mora-Boza, A.; García-Fernández, L. Emerging Biofabrication Techniques: A Review on Natural Polymers for Biomedical Applications. Polymers 2021, 13, 1209. [Google Scholar] [CrossRef] [PubMed]
  4. Tong, X.; Pan, W.; Su, T.; Zhang, M.; Dong, W.; Qi, X. Recent Advances in Natural Polymer-Based Drug Delivery Systems. React. Funct. Polym. 2020, 148, 104501. [Google Scholar] [CrossRef]
  5. Vasile, C.; Pamfil, D.; Stoleru, E.; Baican, M. New Developments in Medical Applications of Hybrid Hydrogels Containing Natural Polymers. Molecules 2020, 25, 1539. [Google Scholar] [CrossRef]
  6. Hamedi, H.; Moradi, S.; Hudson, S.M.; Tonelli, A.E.; King, M.W. Chitosan Based Bioadhesives for Biomedical Applications: A Review. Carbohydr. Polym. 2022, 282, 119100. [Google Scholar] [CrossRef]
  7. Fan, P.; Zeng, Y.; Zaldivar-Silva, D.; Agüero, L.; Wang, S. Chitosan-Based Hemostatic Hydrogels: The Concept, Mechanism, Application, and Prospects. Molecules 2023, 28, 1473. [Google Scholar] [CrossRef] [PubMed]
  8. Ressler, A. Chitosan-Based Biomaterials for Bone Tissue Engineering Applications: A Short Review. Polymers 2022, 14, 3430. [Google Scholar] [CrossRef] [PubMed]
  9. Mohammed, M.H.; Williams, P.A.; Tverezovskaya, O. Extraction of Chitin from Prawn Shells and Conversion to Low Molecular Mass Chitosan. Food Hydrocoll. 2013, 31, 166–171. [Google Scholar] [CrossRef]
  10. Harmsen, R.A.G.; Tuveng, T.R.; Antonsen, S.G.; Eijsink, V.G.H.; Sørlie, M. Can We Make Chitosan by Enzymatic Deacetylation of Chitin? Molecules 2019, 24, 3862. [Google Scholar] [CrossRef]
  11. Kaczmarek, M.B.; Struszczyk-Swita, K.; Li, X.; Szczęsna-Antczak, M.; Daroch, M. Enzymatic Modifications of Chitin, Chitosan, and Chitooligosaccharides. Front. Bioeng. Biotechnol. 2019, 7, 243. [Google Scholar] [CrossRef] [PubMed]
  12. No, H.K.; Meyers, S.P.; Lee, K.S. Isolation and Characterization of Chitin from Crawfish Shell Waste. J. Agric. Food Chem. 1989, 37, 575–579. [Google Scholar] [CrossRef]
  13. Arbia, W.; Arbia, L.; Adour, L.; Amrane, A. Chitin extraction from crustacean shells using biological methods—A review. Food Technol. Biotechnol. 2013, 51, 12–25. [Google Scholar]
  14. Pillai, C.K.S.; Paul, W.; Sharma, C.P. Chitin and Chitosan Polymers: Chemistry, Solubility and Fiber Formation. Prog. Polym. Sci. 2009, 34, 641–678. [Google Scholar] [CrossRef]
  15. Li, X.; Xia, W. Effects of concentration, degree of deacetylation and molecular weight on emulsifying properties of chitosan. Int. J. Biol. Macromol. 2011, 48, 768–772. [Google Scholar] [CrossRef]
  16. 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]
  17. Kou, S.G.; Peters, L.; Mucalo, M. Chitosan: A review of molecular structure, bioactivities and interactions with the human body and micro-organisms. Carbohydr. Polym. 2022, 282, 119132. [Google Scholar] [CrossRef]
  18. Zargar, V.; Asghari, M.; Dashti, A. A Review on Chitin and Chitosan Polymers: Structure, Chemistry, Solubility, Derivatives, and Applications. ChemBioEng Rev. 2015, 2, 204–226. [Google Scholar] [CrossRef]
  19. Kim, S. Competitive Biological Activities of Chitosan and Its Derivatives: Antimicrobial, Antioxidant, Anticancer, and Anti-Inflammatory Activities. Int. J. Polym. Sci. 2018, 2018, 1708172. [Google Scholar] [CrossRef]
  20. Ke, C.-L.; Deng, F.-S.; Chuang, C.-Y.; Lin, C.-H. Antimicrobial Actions and Applications of Chitosan. Polymers 2021, 13, 904. [Google Scholar] [CrossRef] [PubMed]
  21. Negm, N.A.; Hefni, H.H.H.; Abd-Elaal, A.A.A.; Badr, E.A.; Abou Kana, M.T.H. Advancement on Modification of Chitosan Biopolymer and Its Potential Applications. Int. J. Biol. Macromol. 2020, 152, 681–702. [Google Scholar] [CrossRef] [PubMed]
  22. Sahariah, P.; Másson, M. Antimicrobial Chitosan and Chitosan Derivatives: A Review of the Structure–Activity Relationship. Biomacromolecules 2017, 18, 3846–3868. [Google Scholar] [CrossRef]
  23. Argüelles-Monal, W.; Lizardi-Mendoza, J.; Fernández-Quiroz, D.; Recillas-Mota, M.; Montiel-Herrera, M. Chitosan Derivatives: Introducing New Functionalities with a Controlled Molecular Architecture for Innovative Materials. Polymers 2018, 10, 342. [Google Scholar] [CrossRef] [PubMed]
  24. Cohen, E.; Poverenov, E. Hydrophilic Chitosan Derivatives: Synthesis and Applications. Chem. Eur. J. 2022, 28, e202202156. [Google Scholar] [CrossRef]
  25. Boominathan, T.; Sivaramakrishna, A. Recent Advances in the Synthesis, Properties, and Applications of Modified Chitosan Derivatives: Challenges and Opportunities. Top. Curr. Chem. 2021, 379, 19. [Google Scholar] [CrossRef]
  26. Frigaard, J.; Jensen, J.L.; Galtung, H.K.; Hiorth, M. The Potential of Chitosan in Nanomedicine: An Overview of the Cytotoxicity of Chitosan Based Nanoparticles. Front. Pharmacol. 2022, 13, 880377. [Google Scholar] [CrossRef] [PubMed]
  27. Naskar, S.; Kuotsu, K.; Sharma, S. Chitosan-Based Nanoparticles as Drug Delivery Systems: A Review on Two Decades of Research. J. Drug Target. 2019, 27, 379–393. [Google Scholar] [CrossRef]
  28. Precedence Research. Chitosan Market Size to Surpass around US$ 29 Billion by 2030; GlobeNewswire: Los Angeles, CA, USA, 2021. [Google Scholar]
  29. Wang, W.; Meng, Q.; Li, Q.; Liu, J.; Zhou, M.; Jin, Z.; Zhao, K. Chitosan derivatives and their application in biomedicine. Int. J. Mol. Sci. 2020, 21, 487. [Google Scholar] [CrossRef]
  30. Kertmen, A.; Dziedzic, I.; Ehrlich, H. Patentology of chitinous biomaterials. Part II: Chitosan. Carbohydr. Polym. 2023, 301, 120224. [Google Scholar] [CrossRef]
  31. Thambiliyagodage, C.; Jayanetti, M.; Mendis, A.; Ekanayake, G.; Liyanaarachchi, H.; Vigneswaran, S. Recent Advances in Chitosan-Based Applications-A Review. Materials 2023, 16, 2073. [Google Scholar] [CrossRef]
  32. Elangwe, C.N.; Morozkina, S.N.; Olekhnovich, R.O.; Krasichkov, A.; Polyakova, V.O.; Uspenskaya, M.V. A Review on Chitosan and Cellulose Hydrogels for Wound Dressings. Polymers 2022, 14, 5163. [Google Scholar] [CrossRef]
  33. Lacasse, P.; Lanctôt, S.; Fustier, P.; Bégin, A.; Taherian, A.R.; Bisakowski, B. Chitosan hydrogels for accelerating involution and preventing infection of the mammary gland at drying-off. U.S. Patent No. 10, 828, 319, 10 November 2020. [Google Scholar]
  34. Armendariz, I.O.; Gomez, L.E.V.; Estrada, S.A.M. Biodegradable Film Made of Carboximethyl Chitosan and Mimosa Tenuifloraethanolic Extract for Wounds Treatment. Patent MX 2016007284 A, 3 June 2016. Available online: https://lens.org/157-946-545-478-401 (accessed on 4 December 2017).
  35. Travan, A.; Borgogna, M.A.; Parravicini, M.; De Lollis, A.; Siccardi, F. Biocompatible Compositions Comprising a Biocompatible Thickening Polymer and a Chitosan Derivative. U.S. Patent No. 17/597,280, 6 October 2022. [Google Scholar]
  36. Chuan, W.; Xiang, Z.; Lin, S.; Sijing, L.; Mingjuan, J.; Huan, H.; Yongyu, L.; Ting, L.; Yuzhen, Z. Chitosan Oligosaccharide Coated Listeria Iuanuii and Preparation Method and Application Thereof. Patent CN 109248312 A, 23 March 2021. Available online: https://lens.org/019-931-768-976-410 (accessed on 22 January 2019).
  37. Weiwei, W.; Wenshuai, L.; Pingsheng, H.; Huijuan, S.; Deling, K. Double Quaternized Chitosan Derivative, Synthetic Method Thereof, Composite Sponge Biological Dressing Containing Double Quaternized Chitosan Derivative and Application Thereof. Patent CN 111690078 A, 20 September 2022. Available online: https://lens.org/158-377-650-080-779 (accessed on 22 September 2020).
  38. Triviño, G.C. Chitosan Gels (A) Containing Metal Nanoparticles of Copper, Silver and Antibiotics (Ciprofloxacin, Cefotaxime, Gentamicin and Cloxacillin). U.S. Patent No. 16/973,000, 21 October 2021. [Google Scholar]
  39. Alcantar, N.A.; Falahat, R.; Wiranowska, M.; Toomey, R.G.U.S. Enhanced targeted drug delivery system via chitosan hydrogel and chlorotoxin. U.S. Patent No. 9522114B1, 20 December 2016. [Google Scholar]
  40. Aleksandrovna, Z.A.; Vladimirovna, K.M.; Tsyrendorzhievna, S.B.; Viktorovna, S.E.; Petrovich, V.V. Method of Producing Chitosan Derivatives for Visualizing Cell Membranes and Creating Drug Delivery Systems with High Mucoadhesion. Patent RU 2697872 C1, 21 August 2019. Available online: https://lens.org/079-602-098-619-834 (accessed on 27 September 2023).
  41. Jianhua, Z.; Liangxia, W.; Sheng, G.; Ling, C.; Chaoying, W.; Yongli, L.; Wanting, X.; Yumei, M. Chitosan nanoparticle CS-IL-17RC and preparation method thereof. CN 107519481 A, 27 August 2017. Available online: https://lens.org/105-221-674-703-471 (accessed on 29 December 2017).
  42. Panicker, R.K.G.; Veilleux, D.; Lian, P.; Tam, N.C.M.; Fleet, C.; Cheung, A.; Dauphinee, S.; Chen, X.; Jose, L.O.R.A. Chitosan Polyplex-Based Localized Expression of Il-12 Alone or in Combination with Type-I Ifn Inducers for Treatment of Mucosal Cancers. U.S. Patent No. 17/438,922, 24 November 2022. [Google Scholar]
  43. Pillay, V.; Choonara, Y.E.; Kumar, P.; Du Toit, L.C.; Ramburrun, P. Biodegradable Implant. U.S. Patent No. US10478527B2, 19 November 2019. [Google Scholar]
  44. Qiliang, H.; Lidong, C.; Chunli, X.; Chong, C.; Pengyue, Z.; Fengmin, L.; Yongquan, Z. Method for Preparing Prothioconazole Sustained-Release Gel Particles by Carboxymethyl Chitosan-Metal Ion Crosslinking Method. Patent CN 111887244 A, 15 July 2020. Available online: https://lens.org/095-820-269-282-291 (accessed on 6 November 2020).
  45. Arne, T.; Bjugan, A.B.; Linda, H.; Morten, S.; Hafizur, R.; Line, N.A.; Vincent, E. Composition Comprising Chitosan and a Fungicide. U.S. Patent No. 10172353B2, 8 January 2019. [Google Scholar]
  46. Jiajia, C. Berberine and Chitosan Containing Pesticide Composition. Patent CN 110024801 A, 27 May 2019. Available online: https://lens.org/196-874-974-995-823 (accessed on 19 July 2019).
  47. Zhiping, P.; Jichuan, H.; Yuting, T.; Zhijun, L.; Linxiang, Y.; Xuena, W. Method for Preparing Liquid Fertilizer Containing Amino Acid and Chitosan by Using Shrimp-Crab Residual Materials. Patent CN 108456033 A, 2 February 2018. Available online: https://lens.org/040-149-136-430-541 (accessed on 28 August 2018).
  48. Chen, Y.; Li, J.; Li, Q.; Shen, Y.; Ge, Z.; Zhang, W.; Chen, S. Enhanced Water-Solubility, Antibacterial Activity and Biocompatibility upon Introducing Sulfobetaine and Quaternary Ammonium to Chitosan. Carbohydr. Polym. 2016, 143, 246–253. [Google Scholar] [CrossRef]
  49. Ifuku, S.; Matsumoto, C.; Wada, M.; Morimoto, M.; Saimoto, H. Preparation of Highly Regioselective Amphiprotic Chitosan Derivative via “Click Chemistry”. Int. J. Biol. Macromol. 2013, 52, 72–76. [Google Scholar] [CrossRef]
  50. Kurita, K.; Ikeda, H.; Yoshida, Y.; Shimojoh, M.; Harata, M. Chemoselective Protection of the Amino Groups of Chitosan by Controlled Phthaloylation: Facile Preparation of a Precursor Useful for Chemical Modifications. Biomacromolecules 2002, 3, 1–4. [Google Scholar] [CrossRef]
  51. Holappa, J.; Nevalainen, T.; Soininen, P.; Másson, M.; Järvinen, T. Synthesis of Novel Quaternary Chitosan Derivatives via N-Chloroacyl-6-O-Triphenylmethylchitosans. Biomacromolecules 2006, 7, 407–410. [Google Scholar] [CrossRef]
  52. Kurita, K.; Hirakawa, M.; Kikuchi, S.; Yamanaka, H.; Yang, J. Trimethylsilylation of Chitosan and Some Properties of the Product. Carbohydr. Polym. 2004, 56, 333–337. [Google Scholar] [CrossRef]
  53. Rathinam, S.; Ólafsdóttir, S.; Jónsdóttir, S.; Hjálmarsdóttir, M.Á.; Másson, M. Selective Synthesis of N,N,N-Trimethylated Chitosan Derivatives at Different Degree of Substitution and Investigation of Structure-Activity Relationship for Activity against P. Aeruginosa and MRSA. Int. J. Biol. Macromol. 2020, 160, 548–557. [Google Scholar] [CrossRef] [PubMed]
  54. Song, W.; Gaware, V.S.; Rúnarsson, Ö.V.; Másson, M.; Mano, J.F. Functionalized Superhydrophobic Biomimetic Chitosan-Based Films. Carbohydr. Polym. 2010, 81, 140–144. [Google Scholar] [CrossRef]
  55. Sogias, I.A.; Khutoryanskiy, V.V.; Williams, A.C. Exploring the factors affecting the solubility of chitosan in water. Macromol. Chem. Phys. 2010, 211, 426–433. [Google Scholar] [CrossRef]
  56. Panda, P.K.; Yang, J.M.; Chang, Y.H.; Su, W.W. Modification of different molecular weights of chitosan by p-Coumaric acid: Preparation, characterization and effect of molecular weight on its water solubility and antioxidant property. Int. J. Biol. Macromol. 2019, 136, 661–667. [Google Scholar] [CrossRef] [PubMed]
  57. Wu, Q.X.; Lin, D.Q.; Yao, S.J. Design of chitosan and its water soluble derivatives-based drug carriers with polyelectrolyte complexes. Mar. Drugs 2014, 12, 6236–6253. [Google Scholar] [CrossRef]
  58. Panda, P.K.; Dash, P.; Chang, Y.H.; Yang, J.M. Improvement of chitosan water solubility by fumaric acid modification. Mater. Lett. 2022, 316, 132046. [Google Scholar] [CrossRef]
  59. Wu, Q.X.; Lin, D.Q.; Yao, S.J. Fabrication and formation studies on single-walled CA/NaCS-WSC microcapsules. Mater. Sci. Eng. C 2016, 59, 909–915. [Google Scholar] [CrossRef]
  60. Wang, W.; Xue, C.; Mao, X. Chitosan: Structural modification, biological activity and application. Int. J. Biol. Macromol. 2020, 164, 4532–4546. [Google Scholar] [CrossRef] [PubMed]
  61. Vilčnik, A.; Jerman, I.; Šurca Vuk, A.; Koželj, M.; Orel, B.; Tomšič, B.; Simončič, B.; Kovač, J. Structural Properties and Antibacterial Effects of Hydrophobic and Oleophobic Sol−Gel Coatings for Cotton Fabrics. Langmuir 2009, 25, 5869–5880. [Google Scholar] [CrossRef]
  62. Pei, L.; Cai, Z.; Shang, S.; Song, Z. Synthesis and Antibacterial Activity of Alkylated Chitosan under Basic Ionic Liquid Conditions. J. Appl. Polym. Sci. 2014, 131, 40052–40059. [Google Scholar] [CrossRef]
  63. Sun, X.; Li, J.; Shao, K.; Su, C.; Bi, S.; Mu, Y.; Zhang, K.; Cao, Z.; Wang, X.; Chen, X.; et al. A Composite Sponge Based on Alkylated Chitosan and Diatom-Biosilica for Rapid Hemostasis. Int. J. Biol. Macromol. 2021, 182, 2097–2107. [Google Scholar] [CrossRef]
  64. Akopova, T.A.; Demina, T.S.; Cherkaev, G.V.; Khavpachev, M.A.; Bardakova, K.N.; Grachev, A.V.; Vladimirov, L.V.; Zelenetskii, A.N.; Timashev, P.S. Solvent-Free Synthesis and Characterization of Allyl Chitosan Derivatives. RSC Adv. 2019, 9, 20968–20975. [Google Scholar] [CrossRef]
  65. Ma, G.; Yang, D.; Zhou, Y.; Xiao, M.; Kennedy, J.F.; Nie, J. Preparation and Characterization of Water-Soluble N-Alkylated Chitosan. Carbohydr. Polym. 2008, 74, 121–126. [Google Scholar] [CrossRef]
  66. Naidoo, S.; Nomadolo, N.; Matshe, W.M.R.; Cele, Z.; Balogun, M. Exploring the Potential of N-Acylated Chitosan for the Removal of Toxic Pollutants from Wastewater. IOP Conf. Ser. Mater. Sci. Eng. 2019, 655, 012047. [Google Scholar] [CrossRef]
  67. Reis, B.; Gerlach, N.; Steinbach, C.; Haro Carrasco, K.; Oelmann, M.; Schwarz, S.; Müller, M.; Schwarz, D. A Complementary and Revised View on the N-Acylation of Chitosan with Hexanoyl Chloride. Mar. Drugs 2021, 19, 385. [Google Scholar] [CrossRef] [PubMed]
  68. Champagne, L.M. The Synthesis of Water Soluble N-Acyl Chitosan Derivatives for Characterization as Antibacterial Agents; Louisiana State University and Agricultural & Mechanical College: Baton Rouge, LA, USA, 2008. [Google Scholar]
  69. Gamal, A.; Ibrahim, A.G.; Eliwa, E.M.; El-Zomrawy, A.H.; El-Bahy, S.M. Synthesis and Characterization of a Novel Benzothiazole Functionalized Chitosan and Its Use for Effective Adsorption of Cu(II). Int. J. Biol. Macromol. 2021, 183, 1283–1292. [Google Scholar] [CrossRef] [PubMed]
  70. Mohamed, A.E.; Elgammal, W.E.; Eid, A.M.; Dawaba, A.M.; Ibrahim, A.G.; Fouda, A.; Hassan, S.M. Synthesis and Characterization of New Functionalized Chitosan and Its Antimicrobial and In-Vitro Release Behavior from Topical Gel. Int. J. Biol. Macromol. 2022, 207, 242–253. [Google Scholar] [CrossRef]
  71. Ibrahim, A.G.; Fouda, A.; Elgammal, W.E.; Eid, A.M.; Elsenety, M.M.; Mohamed, A.E.; Hassan, S.M. New Thiadiazole Modified Chitosan Derivative to Control the Growth of Human Pathogenic Microbes and Cancer Cell Lines. Sci. Rep. 2022, 12, 21423. [Google Scholar] [CrossRef] [PubMed]
  72. Mohamed, A.E.; Elgammal, W.E.; Dawaba, A.M.; Ibrahim, A.G.; Fouda, A.; Hassan, S.M. A Novel 1,3,4-Thiadiazole Modified Chitosan: Synthesis, Characterization, Antimicrobial Activity, and Release Study from Film Dressings. Appl. Biol. Chem. 2022, 65, 54. [Google Scholar] [CrossRef]
  73. Ibrahim, A.G.; Elgammal, W.E.; Hashem, A.H.; Mohamed, A.E.; Awad, M.A.; Hassan, S.M. Development of a Chitosan Derivative Bearing the Thiadiazole Moiety and Evaluation of Its Antifungal and Larvicidal Efficacy. Polym. Bull. 2023, 1–33. [Google Scholar] [CrossRef]
  74. Deng, P.; Chen, J.; Yao, L.; Zhang, P.; Zhou, J. Thymine-Modified Chitosan with Broad-Spectrum Antimicrobial Activities for Wound Healing. Carbohydr. Polym. 2021, 257, 117630. [Google Scholar] [CrossRef]
  75. Hamodin, A.G.; Elgammal, W.E.; Eid, A.M.; Ibrahim, A.G. Synthesis, Characterization, and Biological Evaluation of New Chitosan Derivative Bearing Diphenyl Pyrazole Moiety. Int. J. Biol. Macromol. 2023, 243, 125180. [Google Scholar] [CrossRef]
  76. Mi, Y.; Li, Q.; Miao, Q.; Tan, W.; Zhang, J.; Guo, Z. Enhanced Antifungal and Antioxidant Activities of New Chitosan Derivatives Modified with Schiff Base Bearing Benzenoid/Heterocyclic Moieties. Int. J. Biol. Macromol. 2022, 208, 586–595. [Google Scholar] [CrossRef]
  77. Wang, L.; Liu, X.; Tan, W.; Li, Q.; Guo, Z.; Zhang, J. Preparation and Antioxidant Activity of Novel Chitosan Oligosaccharide Quinolinyl Urea Derivatives. Carbohydr. Res. 2022, 521, 108678. [Google Scholar] [CrossRef]
  78. Omer, A.M.; Eltaweil, A.S.; El-Fakharany, E.M.; Abd El-Monaem, E.M.; Ismail, M.M.F.; Mohy-Eldin, M.S.; Ayoup, M.S. Novel Cytocompatible Chitosan Schiff Base Derivative as a Potent Antibacterial, Antidiabetic, and Anticancer Agent. Arab. J. Sci. Eng. 2023, 48, 7587–7601. [Google Scholar] [CrossRef]
  79. Antony, R.; Arun, T.; Manickam, S.T.D. A Review on Applications of Chitosan-Based Schiff Bases. Int. J. Biol. Macromol. 2019, 129, 615–633. [Google Scholar] [CrossRef] [PubMed]
  80. Hamed, A.A.; Abdelhamid, I.A.; Saad, G.R.; Elkady, N.A.; Elsabee, M.Z. Synthesis, Characterization and Antimicrobial Activity of a Novel Chitosan Schiff Bases Based on Heterocyclic Moieties. Int. J. Biol. Macromol. 2020, 153, 492–501. [Google Scholar] [CrossRef]
  81. El-Gharably, A.A.; Kenawy, E.-R.S.; Safaan, A.A.; Aboamna, S.A.; Mahmoud, Y.A.-G.; Mandour, H.S.A. Synthesis, Characterization and Application of Chitosan Conjugated Heterocyclic Compounds. J. Polym. Res. 2022, 29, 141. [Google Scholar] [CrossRef]
  82. Hassan, M.A.; Omer, A.M.; Abbas, E.; Baset, W.M.A.; Tamer, T.M. Preparation, Physicochemical Characterization and Antimicrobial Activities of Novel Two Phenolic Chitosan Schiff Base Derivatives. Sci. Rep. 2018, 8, 11416. [Google Scholar] [CrossRef] [PubMed]
  83. Manchaiah, A.S.; Badalamoole, V. Novel Heterocyclic Chitosan-based Schiff Base: Evaluation as Adsorbent for Removal of Methyl Orange from Aqueous Solution. Water Environ. J. 2020, 34, 364–373. [Google Scholar] [CrossRef]
  84. El-Naggar, M.M.; Haneen, D.S.A.; Mehany, A.B.M.; Khalil, M.T. New Synthetic Chitosan Hybrids Bearing Some Heterocyclic Moieties with Potential Activity as Anticancer and Apoptosis Inducers. Int. J. Biol. Macromol. 2020, 150, 1323–1330. [Google Scholar] [CrossRef]
  85. Paula, H.C.B.; Silva, R.B.C.; Santos, C.M.; Dantas, F.D.S.; De Paula, R.C.M.; De Lima, L.R.M.; De Oliveira, E.F.; Figueiredo, E.A.T.; Dias, F.G.B. Eco-Friendly Synthesis of an Alkyl Chitosan Derivative. Int. J. Biol. Macromol. 2020, 163, 1591–1598. [Google Scholar] [CrossRef]
  86. Kritchenkov, A.S.; Egorov, A.R.; Artemjev, A.A.; Kritchenkov, I.S.; Volkova, O.V.; Kiprushkina, E.I.; Zabodalova, L.A.; Suchkova, E.P.; Yagafarov, N.Z.; Tskhovrebov, A.G.; et al. Novel Heterocyclic Chitosan Derivatives and Their Derived Nanoparticles: Catalytic and Antibacterial Properties. Int. J. Biol. Macromol. 2020, 149, 682–692. [Google Scholar] [CrossRef]
  87. Wang, L.; Zhang, J.; Liu, X.; Tan, W.; Li, Q.; Guo, Z. Preparation of Novel Chitosan Oligosaccharide Quaternary Ammonium Derivatives Bearing Quinoline with Antioxidative and Antibacterial Activities. Starch-Stärke 2023, 75, 2300001. [Google Scholar] [CrossRef]
  88. Andreica, B.-I.; Cheng, X.; Marin, L. Quaternary Ammonium Salts of Chitosan. A Critical Overview on the Synthesis and Properties Generated by Quaternization. Eur. Polym. J. 2020, 139, 110016. [Google Scholar] [CrossRef]
  89. Wang, Y.; Yang, Z.; Hu, H.; Wu, J.; Finšgar, M. Indolizine Quaternary Ammonium Salt Inhibitors: The Inhibition and Anti-Corrosion Mechanism of New Dimer Derivatives from Ethyl Acetate Quinolinium Bromide and n-Butyl Quinolinium Bromide. Colloids Surf. Physicochem. Eng. Asp. 2022, 651, 129649. [Google Scholar] [CrossRef]
  90. Muzzarelli, R.A.A.; Tanfani, F. The N-Permethylation of Chitosan and the Preparation of N-Trimethyl Chitosan Iodide. Carbohydr. Polym. 1985, 5, 297–307. [Google Scholar] [CrossRef]
  91. Ali, I.; Nadeem Lone, M.; Al-Othman, Z.A.; Al-Warthan, A.; Marsin Sanagi, M. Heterocyclic Scaffolds: Centrality in Anticancer Drug Development. Curr. Drug Targets 2015, 16, 711–734. [Google Scholar] [CrossRef]
  92. Li, Q.; Wei, L.; Zhang, J.; Gu, G.; Guo, Z. Significantly Enhanced Antioxidant Activity of Chitosan through Chemical Modification with Coumarins. Polym. Chem. 2019, 10, 1480–1488. [Google Scholar] [CrossRef]
  93. Li, Q.; Mi, Y.; Tan, W.; Guo, Z. Highly Efficient Free Radical-Scavenging Property of Phenolic-Functionalized Chitosan Derivatives: Chemical Modification and Activity Assessment. Int. J. Biol. Macromol. 2020, 164, 4279–4288. [Google Scholar] [CrossRef]
  94. Wei, L.; Li, Q.; Chen, Y.; Zhang, J.; Mi, Y.; Dong, F.; Lei, C.; Guo, Z. Enhanced Antioxidant and Antifungal Activity of Chitosan Derivatives Bearing 6-O-Imidazole-Based Quaternary Ammonium Salts. Carbohydr. Polym. 2019, 206, 493–503. [Google Scholar] [CrossRef]
  95. Li, Q.; Zhang, C.; Tan, W.; Gu, G.; Guo, Z. Novel Amino-Pyridine Functionalized Chitosan Quaternary Ammonium Derivatives: Design, Synthesis, and Antioxidant Activity. Molecules 2017, 22, 156. [Google Scholar] [CrossRef]
  96. Wei, L.; Tan, W.; Wang, G.; Li, Q.; Dong, F.; Guo, Z. The Antioxidant and Antifungal Activity of Chitosan Derivatives Bearing Schiff Bases and Quaternary Ammonium Salts. Carbohydr. Polym. 2019, 226, 115256. [Google Scholar] [CrossRef]
  97. Zhang, J.; Tan, W.; Li, Q.; Dong, F.; Guo, Z. Synthesis and Characterization of N,N,N-Trimethyl-O-(Ureidopyridinium)Acetyl Chitosan Derivatives with Antioxidant and Antifungal Activities. Mar. Drugs 2020, 18, 163. [Google Scholar] [CrossRef] [PubMed]
  98. Zhang, J.; Mi, Y.; Sun, X.; Chen, Y.; Miao, Q.; Tan, W.; Li, Q.; Dong, F.; Guo, Z. Improved Antioxidant and Antifungal Activity of Chitosan Derivatives Bearing Urea Groups. Starch-Stärke 2020, 72, 1900205. [Google Scholar] [CrossRef]
  99. Wei, L.; Mi, Y.; Zhang, J.; Li, Q.; Dong, F.; Guo, Z. Evaluation of Quaternary Ammonium Chitosan Derivatives Differing in the Length of Alkyl Side-Chain: Synthesis and Antifungal Activity. Int. J. Biol. Macromol. 2019, 129, 1127–1132. [Google Scholar] [CrossRef] [PubMed]
  100. Jiang, Y.; Zhang, Y.; Zhang, H.; Zhu, R.; Hu, Y. Synthesis of N-alkylated Quaternary Ammonium Chitosan and Its Long-term Antibacterial Finish for Rabbit Hair Fabric. Polym. Adv. Technol. 2022, 33, 314–325. [Google Scholar] [CrossRef]
  101. Mi, Y.; Zhang, J.; Han, X.; Tan, W.; Miao, Q.; Cui, J.; Li, Q.; Guo, Z. Modification of Carboxymethyl Inulin with Heterocyclic Compounds: Synthesis, Characterization, Antioxidant and Antifungal Activities. Int. J. Biol. Macromol. 2021, 181, 572–581. [Google Scholar] [CrossRef]
  102. Mi, Y.; Miao, Q.; Cui, J.; Tan, W.; Guo, Z. Novel 2-Hydroxypropyltrimethyl Ammonium Chitosan Derivatives: Synthesis, Characterization, Moisture Absorption and Retention Properties. Molecules 2021, 26, 4238. [Google Scholar] [CrossRef] [PubMed]
  103. Li, W.; Duan, Y.; Huang, J.; Zheng, Q. Synthesis, Antioxidant and Cathepsin D Inhibition Activity of Quaternary Ammonium Chitosan Derivatives. Carbohydr. Polym. 2016, 136, 884–891. [Google Scholar] [CrossRef] [PubMed]
  104. Spinelli, V.A.; Laranjeira, M.C.M.; Fávere, V.T. Preparation and Characterization of Quaternary Chitosan Salt: Adsorption Equilibrium of Chromium(VI) Ion. React. Funct. Polym. 2004, 61, 347–352. [Google Scholar] [CrossRef]
  105. Sangeetha, Y.; Meenakshi, S.; SairamSundaram, C. Corrosion Mitigation of N-(2-Hydroxy-3-Trimethyl Ammonium)Propyl Chitosan Chloride as Inhibitor on Mild Steel. Int. J. Biol. Macromol. 2015, 72, 1244–1249. [Google Scholar] [CrossRef]
  106. Tan, W.; Lin, C.; Zhang, J.; Li, Q.; Guo, Z. Synthesis of Hydroxypropyltrimethyl Ammonium Chitosan Derivatives Bearing Thioctate and the Potential for Antioxidant Application. Molecules 2022, 27, 2682. [Google Scholar] [CrossRef]
  107. Li, P.; He, L.; Li, X.; Liu, X.; Sun, M. Corrosion Inhibition Effect of N-(4-Diethylaminobenzyl) Quaternary Ammonium Chitosan for X80 Pipeline Steel in Hydrochloric Acid Solution. Int. J. Electrochem. Sci. 2021, 16, 150931. [Google Scholar] [CrossRef]
  108. Li, Z.; Yang, F.; Yang, R. Synthesis and Characterization of Chitosan Derivatives with Dual-Antibacterial Functional Groups. Int. J. Biol. Macromol. 2015, 75, 378–387. [Google Scholar] [CrossRef] [PubMed]
  109. Kratky, M.; Vinsova, J. Antimycobacterial activity of quaternary pyridinium salts and pyridinium N-oxides-review. Curr. Pharm. Des. 2013, 19, 1343–1355. [Google Scholar] [CrossRef] [PubMed]
  110. Sowmiah, S.; Esperança, J.M.S.S.; Rebelo, L.P.N.; Afonso, C.A.M. Pyridinium Salts: From Synthesis to Reactivity and Applications. Org. Chem. Front. 2018, 5, 453–493. [Google Scholar] [CrossRef]
  111. Wei, L.; Li, Q.; Tan, W.; Dong, F.; Luan, F.; Guo, Z. Synthesis, Characterization, and the Antioxidant Activity of Double Quaternized Chitosan Derivatives. Molecules 2017, 22, 501. [Google Scholar] [CrossRef] [PubMed]
  112. Oyervides-Muñoz, E.; Avérous, L.; Sosa-Santillán, G.D.J.; Pollet, E.; Pérez-Aguilar, N.V.; Rojas-Caldera, C.M.; Fuentes-Avilés, J.G.; García-Astrain, C. EDC-Mediated Grafting of Quaternary Ammonium Salts onto Chitosan for Antibacterial and Thermal Properties Improvement. Macromol. Chem. Phys. 2019, 220, 1800530. [Google Scholar] [CrossRef]
  113. Franconetti, A.; Contreras-Bernal, L.; Prado-Gotor, R.; Cabrera-Escribano, F. Synthesis of Hyperpolarizable Biomaterials at Molecular Level Based on Pyridinium–Chitosan Complexes. RSC Adv. 2015, 5, 74274–74283. [Google Scholar] [CrossRef]
  114. Jia, R.; Duan, Y.; Fang, Q.; Wang, X.; Huang, J. Pyridine-Grafted Chitosan Derivative as an Antifungal Agent. Food Chem. 2016, 196, 381–387. [Google Scholar] [CrossRef]
  115. Lin, C.; Guo, Z.; Jiang, A.; Liang, X.; Tan, W. Cationic Chitooligosaccharide Derivatives Bearing Pyridinium and Trialkyl Ammonium: Preparation, Characterization and Antimicrobial Activities. Polymers 2022, 15, 14. [Google Scholar] [CrossRef]
  116. Zhang, J.; Zhang, S.; Wang, L.; Tan, W.; Li, Q.; Guo, Z. The Antioxidant and Antibacterial Activities of the Pyridine-4-Aldehyde Schiff Bases Grafted Chloracetyl Chitosan Oligosaccharide Derivatives. Starch-Stärke 2023, 75, 2100268. [Google Scholar] [CrossRef]
  117. Zhang, J.; Sun, X.; Chen, Y.; Mi, Y.; Tan, W.; Miao, Q.; Li, Q.; Dong, F.; Guo, Z. Preparation of 2,6-Diurea-Chitosan Oligosaccharide Derivatives for Efficient Antifungal and Antioxidant Activities. Carbohydr. Polym. 2020, 234, 115903. [Google Scholar] [CrossRef]
  118. Zincke, T.; Heuser, G.; Möller, W.I. Ueber Dinitrophenylpyridiniumchlorid und Dessen Umwandlungsproducte. Justus Liebigs Ann. Chem. 1904, 333, 296–345. [Google Scholar] [CrossRef]
  119. Cheng, W.-C.; Kurth, M.J. The Zincke Reaction. A Review. Org. Prep. Proced. Int. 2002, 34, 585–608. [Google Scholar] [CrossRef]
  120. Venegas-García, D.J.; Steiger, B.G.K.; Wilson, L.D. A Pyridinium-Modified Chitosan-Based Adsorbent for Arsenic Removal via a Coagulation-like Methodology. RSC Sustain. 2023, 1, 1259–1269. [Google Scholar] [CrossRef]
  121. Gonçalves, F.; Freitas, R. Modification of Chitosan by Zincke Reaction: Synthesis of a Novel Polycationic Chitosan-Pyridinium Derivative. J. Braz. Chem. Soc. 2019, 30, 2318–2323. [Google Scholar] [CrossRef]
  122. Meng, X.; Edgar, K.J. “Click” Reactions in Polysaccharide Modification. Prog. Polym. Sci. 2016, 53, 52–85. [Google Scholar] [CrossRef]
  123. Li, Q.; Sun, X.; Gu, G.; Guo, Z. Novel Water Soluble Chitosan Derivatives with 1,2,3-Triazolium and Their Free Radical-Scavenging Activity. Mar. Drugs 2018, 16, 107. [Google Scholar] [CrossRef]
  124. Tan, W.; Zhang, J.; Mi, Y.; Dong, F.; Li, Q.; Guo, Z. Synthesis, Characterization, and Evaluation of Antifungal and Antioxidant Properties of Cationic Chitosan Derivative via Azide-Alkyne Click Reaction. Int. J. Biol. Macromol. 2018, 120, 318–324. [Google Scholar] [CrossRef]
  125. Tan, W.; Zhang, J.; Mi, Y.; Dong, F.; Li, Q.; Guo, Z. Enhanced Antifungal Activity of Novel Cationic Chitosan Derivative Bearing Triphenylphosphonium Salt via Azide-Alkyne Click Reaction. Int. J. Biol. Macromol. 2020, 165, 1765–1772. [Google Scholar] [CrossRef]
  126. Kim, S.-H.; Semenya, D.; Castagnolo, D. Antimicrobial Drugs Bearing Guanidine Moieties: A Review. Eur. J. Med. Chem. 2021, 216, 113293. [Google Scholar] [CrossRef]
  127. Eckert-Maksić, M.; Glasovac, Z.; Trošelj, P.; Kütt, A.; Rodima, T.; Koppel, I.; Koppel, I.A. Basicity of Guanidines with Heteroalkyl Side Chains in Acetonitrile. Eur. J. Org. Chem. 2008, 2008, 5176–5184. [Google Scholar] [CrossRef]
  128. Zhai, X.; Sun, P.; Luo, Y.; Ma, C.; Xu, J.; Liu, W. Guanidinylation: A Simple Way to Fabricate Cell Penetrating Peptide Analogue-Modified Chitosan Vector for Enhanced Gene Delivery: Guanidinylation. J. Appl. Polym. Sci. 2011, 121, 3569–3578. [Google Scholar] [CrossRef]
  129. Hu, Y.; Du, Y.; Yang, J.; Kennedy, J.; Wang, X.; Wang, L. Synthesis, Characterization and Antibacterial Activity of Guanidinylated Chitosan. Carbohydr. Polym. 2007, 67, 66–72. [Google Scholar] [CrossRef]
  130. Lahmer, R.A.; Williams, A.P.; Townsend, S.; Baker, S.; Jones, D.L. Antibacterial Action of Chitosan-Arginine against Escherichia Coli O157 in Chicken Juice. Food Control 2012, 26, 206–211. [Google Scholar] [CrossRef]
  131. Lv, J.; Jiang, M.; Fang, Y.; Zhang, W.; Wu, M.; Zheng, F.; Lei, K.; Shang, L.; Zhao, Y. The Mimicked Cell Penetration Peptides of Chitosan Derivates with Low Molecular Weight for Transdermal Enhancement Studies. Eur. Polym. J. 2023, 195, 112197. [Google Scholar] [CrossRef]
  132. Zhou, Y.; Liu, G.; Huang, H.; Wu, J. Advances and Impact of Arginine-Based Materials in Wound Healing. J. Mater. Chem. B 2021, 9, 6738–6750. [Google Scholar] [CrossRef] [PubMed]
  133. Xiao, B.; Wan, Y.; Zhao, M.; Liu, Y.; Zhang, S. Preparation and Characterization of Antimicrobial Chitosan-N-Arginine with Different Degrees of Substitution. Carbohydr. Polym. 2011, 83, 144–150. [Google Scholar] [CrossRef]
  134. Lv, H.-X.; Zhang, Z.-H.; Wang, X.-P.; Cheng, Q.-Q.; Wang, W.; Huang, X.-H.; Zhou, J.-P.; Zhang, Q.; Hou, L.-L.; Huo, W. A Biomimetic Chitosan Derivates: Preparation, Characterization and Transdermal Enhancement Studies of N-Arginine Chitosan. Molecules 2011, 16, 6778–6790. [Google Scholar] [CrossRef]
  135. Cui, J.; Sun, Y.; Wang, L.; Miao, Q.; Tan, W.; Guo, Z. Preparation of L-Arginine Schiff Bases Modified Chitosan Derivatives and Their Antimicrobial and Antioxidant Properties. Mar. Drugs 2022, 20, 688. [Google Scholar] [CrossRef]
  136. Salama, H.E.; Saad, G.R.; Sabaa, M.W. Synthesis, Characterization and Antimicrobial Activity of Biguanidinylated Chitosan-g-Poly[(R)-3-Hydroxybutyrate]. Int. J. Biol. Macromol. 2017, 101, 438–447. [Google Scholar] [CrossRef] [PubMed]
  137. Sang, W.; Tang, Z.; He, M.-Y.; Hua, Y.-P.; Xu, Q. Synthesis and Preservative Application of Quaternized Carboxymethyl Chitosan Containing Guanidine Groups. Int. J. Biol. Macromol. 2015, 75, 489–494. [Google Scholar] [CrossRef]
  138. Sahariah, P.; Óskarsson, B.M.; Hjálmarsdóttir, M.Á.; Másson, M. Synthesis of Guanidinylated Chitosan with the Aid of Multiple Protecting Groups and Investigation of Antibacterial Activity. Carbohydr. Polym. 2015, 127, 407–417. [Google Scholar] [CrossRef] [PubMed]
  139. Chen, S.; Li, C.; Hou, T.; Cai, Y.; Liang, L.; Chen, L.; Li, M. Polyhexamethylene Guanidine Functionalized Chitosan Nanofiber Membrane with Superior Adsorption and Antibacterial Performances. React. Funct. Polym. 2019, 145, 104379. [Google Scholar] [CrossRef]
  140. Salama, A.; Hesemann, P. New N-Guanidinium Chitosan/Silica Ionic Microhybrids as Efficient Adsorbent for Dye Removal from Waste Water. Int. J. Biol. Macromol. 2018, 111, 762–768. [Google Scholar] [CrossRef] [PubMed]
  141. Salama, A.; Hasanin, M.; Hesemann, P. Synthesis and Antimicrobial Properties of New Chitosan Derivatives Containing Guanidinium Groups. Carbohydr. Polym. 2020, 241, 116363. [Google Scholar] [CrossRef] [PubMed]
  142. Shaabani, A.; Sedghi, R. Preparation of Chitosan Biguanidine/PANI-Containing Self-Healing Semi-Conductive Waterborne Scaffolds for Bone Tissue Engineering. Carbohydr. Polym. 2021, 264, 118045. [Google Scholar] [CrossRef] [PubMed]
  143. Agrahari, A.K.; Bose, P.; Jaiswal, M.K.; Rajkhowa, S.; Singh, A.S.; Hotha, S.; Mishra, N.; Tiwari, V.K. Cu(I)-Catalyzed Click Chemistry in Glycoscience and Their Diverse Applications. Chem. Rev. 2021, 121, 7638–7956. [Google Scholar] [CrossRef]
  144. Badria, A. Click Chemistry: A Promising Tool for Building Hierarchical Structures. Polymers 2022, 14, 4077. [Google Scholar] [CrossRef] [PubMed]
  145. Chen, Y.; Yu, L.; Zhang, B.; Feng, W.; Xu, M.; Gao, L.; Liu, N.; Wang, Q.; Huang, X.; Li, P.; et al. Design and Synthesis of Biocompatible, Hemocompatible, and Highly Selective Antimicrobial Cationic Peptidopolysaccharides via Click Chemistry. Biomacromolecules 2019, 20, 2230–2240. [Google Scholar] [CrossRef]
  146. Rathinam, S.; Hjálmarsdóttir, M.Á.; Thygesen, M.B.; Másson, M. Chitotriazolan (Poly(β(1-4)-2-(1H-1,2,3-Triazol-1-Yl)-2-Deoxy-d-Glucose)) Derivatives: Synthesis, Characterization, and Evaluation of Antibacterial Activity. Carbohydr. Polym. 2021, 267, 118162. [Google Scholar] [CrossRef]
  147. Chen, Y.; Wang, F.; Yun, D.; Guo, Y.; Ye, Y.; Wang, Y.; Tan, H. Preparation of a C6 Quaternary Ammonium Chitosan Derivative through a Chitosan Schiff Base with Click Chemistry. J. Appl. Polym. Sci. 2013, 129, 3185–3191. [Google Scholar] [CrossRef]
  148. Tirino, P.; Laurino, R.; Maglio, G.; Malinconico, M.; d’Ayala, G.G.; Laurienzo, P. Synthesis of Chitosan–PEO Hydrogels via Mesylation and Regioselective Cu(I)-Catalyzed Cycloaddition. Carbohydr. Polym. 2014, 112, 736–745. [Google Scholar] [CrossRef] [PubMed]
  149. Koshiji, K.; Nonaka, Y.; Iwamura, M.; Dai, F.; Matsuoka, R.; Hasegawa, T. C6-Modifications on Chitosan to Develop Chitosan-Based Glycopolymers and Their Lectin-Affinities with Sigmoidal Binding Profiles. Carbohydr. Polym. 2016, 137, 277–286. [Google Scholar] [CrossRef] [PubMed]
  150. Barbosa, M.; Vale, N.; Costa, F.M.T.A.; Martins, M.C.L.; Gomes, P. Tethering Antimicrobial Peptides onto Chitosan: Optimization of Azide-Alkyne “Click” Reaction Conditions. Carbohydr. Polym. 2017, 165, 384–393. [Google Scholar] [CrossRef]
  151. Qin, Y.; Liu, S.; Xing, R.; Li, K.; Yu, H.; Li, P. Synthesis and Antifungal Evaluation of (1,2,3-Triazol-4-Yl)Methyl Nicotinate Chitosan. Int. J. Biol. Macromol. 2013, 61, 58–62. [Google Scholar] [CrossRef]
  152. Tan, W.; Zhang, J.; Zhao, X.; Dong, F.; Li, Q.; Guo, Z. Synthesis and Antioxidant Action of Chitosan Derivatives with Amino-Containing Groups via Azide-Alkyne Click Reaction and N-Methylation. Carbohydr. Polym. 2018, 199, 583–592. [Google Scholar] [CrossRef]
  153. Lunkov, A.; Shagdarova, B.; Lyalina, T.; Dubinnyi, M.A.; Karpova, N.; Lopatin, S.; Il’ina, A.; Varlamov, V. Simple Method for Ultrasound Assisted «click» Modification of Azido-Chitosan Derivatives by CuAAC. Carbohydr. Polym. 2022, 282, 119109. [Google Scholar] [CrossRef]
  154. Kritchenkov, A.S.; Egorov, A.R.; Dysin, A.P.; Volkova, O.V.; Zabodalova, L.A.; Suchkova, E.P.; Kurliuk, A.V.; Shakola, T.V. Ultrasound-Assisted Cu(I)-Catalyzed Azide-Alkyne Click Cycloaddition as Polymer-Analogous Transformation in Chitosan Chemistry. High Antibacterial and Transfection Activity of Novel Triazol Betaine Chitosan Derivatives and Their Nanoparti-cles. Int. J. Biol. Macromol. 2019, 137, 592–603. [Google Scholar] [CrossRef]
  155. Sahariah, P.; Másson, M. Efficient Synthesis of Chitosan Derivatives as Clickable Tools. Eur. Polym. J. 2022, 166, 111039. [Google Scholar] [CrossRef]
  156. Nornberg, A.B.; De Aquino, T.F.B.; Martins, C.C.; Luchese, C.; Wilhelm, E.A.; Jacob, R.G.; Hartwig, D.; Fajardo, A.R. Organoselenium-Chitosan Derivative: Synthesis via “Click” Reaction, Characterization and Antioxidant Activity. Int. J. Biol. Macromol. 2021, 191, 19–26. [Google Scholar] [CrossRef]
  157. Manisha D., P.; Chawla, R.; Dutta, P.K. “Click” Synthesized Non-Substituted Triazole Modified Chitosan from CaC2 as a Novel Antibacterial and Antioxidant Polymer. J. Polym. Res. 2022, 29, 179. [Google Scholar] [CrossRef]
  158. Guaresti, O.; García–Astrain, C.; Palomares, T.; Alonso–Varona, A.; Eceiza, A.; Gabilondo, N. Synthesis and Characterization of a Biocompatible Chitosan–Based Hydrogel Cross–Linked via ‘Click’ Chemistry for Controlled Drug Release. Int. J. Biol. Macromol. 2017, 102, 1–9. [Google Scholar] [CrossRef] [PubMed]
  159. Ruiz-Pardo, C.; Silva-Gutiérrez, L.; Lizardi-Mendoza, J.; López-Franco, Y.; Peniche-Covas, C.; Argüelles-Monal, W. Chitosan Hydrogels Based on the Diels–Alder Click Reaction: Rheological and Kinetic Study. Polymers 2022, 14, 1202. [Google Scholar] [CrossRef] [PubMed]
  160. Azmy, E.A.M.; Hashem, H.E.; Mohamed, E.A.; Negm, N.A. Synthesis, Characterization, Swelling and Antimicrobial Efficacies of Chemically Modified Chitosan Biopolymer. J. Mol. Liq. 2019, 284, 748–754. [Google Scholar] [CrossRef]
  161. Muzzarelli, R.; El Mehtedi, M.; Bottegoni, C.; Aquili, A.; Gigante, A. Genipin-Crosslinked Chitosan Gels and Scaffolds for Tissue Engineering and Regeneration of Cartilage and Bone. Mar. Drugs 2015, 13, 7314–7338. [Google Scholar] [CrossRef]
  162. Kandile, N.G.; Mohamed, H.M. New Chitosan Derivatives Inspired on Heterocyclic Anhydride of Potential Bioactive for Medical Applications. Int. J. Biol. Macromol. 2021, 182, 1543–1553. [Google Scholar] [CrossRef]
  163. Kandile, N.G.; Mohamed, M.I.; Zaky, H.T.; Nasr, A.S.; Ali, Y.G. Quinoline Anhydride Derivatives Cross-Linked Chitosan Hydrogels for Potential Use in Biomedical and Metal Ions Adsorption. Polym. Bull. 2022, 79, 2461–2486. [Google Scholar] [CrossRef]
  164. Rabea, E.I.; Badawy, M.E.-T.; Stevens, C.V.; Smagghe, G.; Steurbaut, W. Chitosan as Antimicrobial Agent: Applications and Mode of Action. Biomacromolecules 2003, 4, 1457–1465. [Google Scholar] [CrossRef]
  165. Zia, K.M.; Barikani, M.; Zuber, M.; Bhatti, I.A.; Sheikh, M.A. Molecular engineering of chitin based polyurethane elastomers. Carbohy. Polym. 2008, 74, 149–158. [Google Scholar] [CrossRef]
  166. Sudarshan, N.R.; Hoover, D.G.; Knorr, D. Antibacterial action of chitosan. Food Biotechnol. 1992, 6, 257–272. [Google Scholar] [CrossRef]
  167. Perinelli, D.R.; Fagioli, L.; Campana, R.; Lam, J.K.W.; Baffone, W.; Palmieri, G.F.; Casettari, L.; Bonacucina, G. Chitosan-Based Nanosystems and Their Exploited Antimicrobial Activity. Eur. J. Pharm. Sci. 2018, 117, 8–20. [Google Scholar] [CrossRef] [PubMed]
  168. Yilmaz Atay, H. Antibacterial Activity of Chitosan-Based Systems. In Functional Chitosan; Jana, S., Jana, S., Eds.; Springer: Singapore, 2019; pp. 457–489. ISBN 9789811502620. [Google Scholar]
  169. Kong, M.; Chen, X.G.; Xing, K.; Park, H.J. Antimicrobial Properties of Chitosan and Mode of Action: A State of the Art Review. Int. J. Food Microbiol. 2010, 144, 51–63. [Google Scholar] [CrossRef] [PubMed]
  170. Jeon, Y. Antimicrobial Effect of Chitooligosaccharides Produced by Bioreactor. Carbohydr. Polym. 2001, 44, 71–76. [Google Scholar] [CrossRef]
  171. Xing, K.; Chen, X.G.; Liu, C.S.; Cha, D.S.; Park, H.J. Oleoyl-Chitosan Nanoparticles Inhibits Escherichia Coli and Staphylococcus Aureus by Damaging the Cell Membrane and Putative Binding to Extracellular or Intracellular Targets. Int. J. Food Microbiol. 2009, 132, 127–133. [Google Scholar] [CrossRef] [PubMed]
  172. Clifton, L.A.; Skoda, M.W.A.; Le Brun, A.P.; Ciesielski, F.; Kuzmenko, I.; Holt, S.A.; Lakey, J.H. Effect of Divalent Cation Removal on the Structure of Gram-Negative Bacterial Outer Membrane Models. Langmuir 2015, 31, 404–412. [Google Scholar] [CrossRef]
  173. Hosseinnejad, M.; Jafari, S.M. Evaluation of Different Factors Affecting Antimicrobial Properties of Chitosan. Int. J. Biol. Macromol. 2016, 85, 467–475. [Google Scholar] [CrossRef]
  174. Brown, S.; Santa Maria, J.P.; Walker, S. Wall Teichoic Acids of Gram-Positive Bacteria. Annu. Rev. Microbiol. 2013, 67, 313–336. [Google Scholar] [CrossRef] [PubMed]
  175. Pasquina-Lemonche, L.; Burns, J.; Turner, R.D.; Kumar, S.; Tank, R.; Mullin, N.; Wilson, J.S.; Chakrabarti, B.; Bullough, P.A.; Foster, S.J.; et al. The Architecture of the Gram-Positive Bacterial Cell Wall. Nature 2020, 582, 294–297. [Google Scholar] [CrossRef]
  176. Matica, M.A.; Aachmann, F.L.; Tøndervik, A.; Sletta, H.; Ostafe, V. Chitosan as a Wound Dressing Starting Material: Antimicrobial Properties and Mode of Action. Int. J. Mol. Sci. 2019, 20, 5889. [Google Scholar] [CrossRef]
  177. Je, J.-Y.; Kim, S.-K. Chitosan Derivatives Killed Bacteria by Disrupting the Outer and Inner Membrane. J. Agric. Food Chem. 2006, 54, 6629–6633. [Google Scholar] [CrossRef]
  178. Feng, P.; Luo, Y.; Ke, C.; Qiu, H.; Wang, W.; Zhu, Y.; Hou, R.; Xu, L.; Wu, S. Chitosan-Based Functional Materials for Skin Wound Repair: Mechanisms and Applications. Front. Bioeng. Biotechnol. 2021, 9, 650598. [Google Scholar] [CrossRef] [PubMed]
  179. Gan, L.; Chen, S.; Jensen, G.J. Molecular Organization of Gram-Negative Peptidoglycan. Proc. Natl. Acad. Sci. USA 2008, 105, 18953–18957. [Google Scholar] [CrossRef]
  180. Khalid, S.; Piggot, T.J.; Samsudin, F. Atomistic and Coarse Grain Simulations of the Cell Envelope of Gram-Negative Bacteria: What Have We Learned? Acc. Chem. Res. 2019, 52, 180–188. [Google Scholar] [CrossRef] [PubMed]
  181. Helander, I.M.; Nurmiaho-Lassila, E.-L.; Ahvenainen, R.; Rhoades, J.; Roller, S. Chitosan Disrupts the Barrier Properties of the Outer Membrane of Gram-Negative Bacteria. Int. J. Food Microbiol. 2001, 71, 235–244. [Google Scholar] [CrossRef] [PubMed]
  182. Chen, C.-P.; Hsieh, C.-M.; Tsai, T.; Yang, J.-C.; Chen, C.-T. Optimization and Evaluation of a Chitosan/Hydroxypropyl Methylcellulose Hydrogel Containing Toluidine Blue O for Antimicrobial Photodynamic Inactivation. Int. J. Mol. Sci. 2015, 16, 20859–20872. [Google Scholar] [CrossRef]
  183. Xia, W.; Wei, X.-Y.; Xie, Y.-Y.; Zhou, T. A Novel Chitosan Oligosaccharide Derivative: Synthesis, Antioxidant and Antibacterial Properties. Carbohydr. Polym. 2022, 291, 119608. [Google Scholar] [CrossRef]
  184. Arjunan, N.; Kumari, H.L.J.; Singaravelu, C.M.; Kandasamy, R.; Kandasamy, J. Physicochemical Investigations of Biogenic Chitosan-Silver Nanocomposite as Antimicrobial and Anticancer Agent. Int. J. Biol. Macromol. 2016, 92, 77–87. [Google Scholar] [CrossRef]
  185. Rahayu, D.P.; De Mori, A.; Yusuf, R.; Draheim, R.; Lalatsa, A.; Roldo, M. Enhancing the Antibacterial Effect of Chitosan to Combat Orthopaedic Implant-Associated Infections. Carbohydr. Polym. 2022, 289, 119385. [Google Scholar] [CrossRef]
  186. Tamer, T.M.; Zhou, H.; Hassan, M.A.; Abu-Serie, M.M.; Shityakov, S.; Elbayomi, S.M.; Mohy-Eldin, M.S.; Zhang, Y.; Cheang, T. Synthesis and Physicochemical Properties of an Aromatic Chitosan Derivative: In Vitro Antibacterial, Antioxidant, and Anticancer Evaluations, and in Silico Studies. Int. J. Biol. Macromol. 2023, 240, 124339. [Google Scholar] [CrossRef]
  187. Hassan, M.A.; Tamer, T.M.; Omer, A.M.; Baset, W.M.A.; Abbas, E.; Mohy-Eldin, M.S. Therapeutic Potential of Two Formulated Novel Chitosan Derivatives with Prominent Antimicrobial Activities against Virulent Microorganisms and Safe Profiles toward Fibroblast Cells. Int. J. Pharm. 2023, 634, 122649. [Google Scholar] [CrossRef]
  188. Zaki, N.M.; Hafez, M.M. Enhanced Antibacterial Effect of Ceftriaxone Sodium-Loaded Chitosan Nanoparticles Against Intracellular Salmonella Typhimurium. AAPS PharmSciTech 2012, 13, 411–421. [Google Scholar] [CrossRef] [PubMed]
  189. Jamil, B.; Habib, H.; Abbasi, S.; Nasir, H.; Rahman, A.; Rehman, A.; Bokhari, H.; Imran, M. Cefazolin Loaded Chitosan Nanoparticles to Cure Multi Drug Resistant Gram-Negative Pathogens. Carbohydr. Polym. 2016, 136, 682–691. [Google Scholar] [CrossRef]
  190. Maya, S.; Indulekha, S.; Sukhithasri, V.; Smitha, K.T.; Nair, S.V.; Jayakumar, R.; Biswas, R. Efficacy of Tetracycline Encapsulated O-Carboxymethyl Chitosan Nanoparticles against Intracellular Infections of Staphylococcus Aureus. Int. J. Biol. Macromol. 2012, 51, 392–399. [Google Scholar] [CrossRef]
  191. Mawad, A.; Helmy, Y.A.; Shalkami, A.-G.; Kathayat, D.; Rajashekara, G.E. Coli Nissle Microencapsulation in Alginate-Chitosan Nanoparticles and Its Effect on Campylobacter Jejuni in Vitro. Appl. Microbiol. Biotechnol. 2018, 102, 10675–10690. [Google Scholar] [CrossRef] [PubMed]
  192. Rhim, J.-W.; Hong, S.-I.; Park, H.-M.; Ng, P.K.W. Preparation and Characterization of Chitosan-Based Nanocomposite Films with Antimicrobial Activity. J. Agric. Food Chem. 2006, 54, 5814–5822. [Google Scholar] [CrossRef]
  193. Kumar-Krishnan, S.; Prokhorov, E.; Hernández-Iturriaga, M.; Mota-Morales, J.D.; Vázquez-Lepe, M.; Kovalenko, Y.; Sanchez, I.C.; Luna-Bárcenas, G. Chitosan/Silver Nanocomposites: Synergistic Antibacterial Action of Silver Nanoparticles and Silver Ions. Eur. Polym. J. 2015, 67, 242–251. [Google Scholar] [CrossRef]
  194. Mu, H.; Liu, Q.; Niu, H.; Sun, Y.; Duan, J. Gold Nanoparticles Make Chitosan–Streptomycin Conjugates Effective towards Gram-Negative Bacterial Biofilm. RSC Adv. 2016, 6, 8714–8721. [Google Scholar] [CrossRef]
  195. P, M.R.; Muraleedaran, K.; Mujeeb, V.M.A. Applications of Chitosan Powder with in Situ Synthesized Nano ZnO Particles as an Antimicrobial Agent. Int. J. Biol. Macromol. 2015, 77, 266–272. [Google Scholar] [CrossRef] [PubMed]
  196. Hanpanich, O.; Wongkongkatep, P.; Pongtharangkul, T.; Wongkongkatep, J. Turning Hydrophilic Bacteria into Biorenewable Hydrophobic Material with Potential Antimicrobial Activity via Interaction with Chitosan. Bioresour. Technol. 2017, 230, 97–102. [Google Scholar] [CrossRef]
  197. Rivera Aguayo, P.; Bruna Larenas, T.; Alarcón Godoy, C.; Cayupe Rivas, B.; González-Casanova, J.; Rojas-Gómez, D.; Caro Fuentes, N. Antimicrobial and Antibiofilm Capacity of Chitosan Nanoparticles against Wild Type Strain of Pseudomonas sp. Isolated from Milk of Cows Diagnosed with Bovine Mastitis. Antibiotics 2020, 9, 551. [Google Scholar] [CrossRef]
  198. Muñoz-Nuñez, C.; Cuervo-Rodríguez, R.; Echeverría, C.; Fernández-García, M.; Muñoz-Bonilla, A. Synthesis and Characterization of Thiazolium Chitosan Derivative with Enhanced Antimicrobial Properties and Its Use as Component of Chitosan Based Films. Carbohydr. Polym. 2023, 302, 120438. [Google Scholar] [CrossRef]
  199. Zou, W.; Gu, J.; Li, J.; Wang, Y.; Chen, S. Tailorable Antibacterial and Cytotoxic Chitosan Derivatives by Introducing Quaternary Ammonium Salt and Sulfobetaine. Int. J. Biol. Macromol. 2022, 218, 992–1001. [Google Scholar] [CrossRef]
  200. Alenzi, A.M.; Albalawi, S.A.; Alghamdi, S.G.; Albalawi, R.F.; Albalawi, H.S.; Qushawy, M. Review on Different Vesicular Drug Delivery Systems (VDDSs) and TheirApplications. Recent Pat. Nanotechnol. 2023, 17, 18–32. [Google Scholar] [CrossRef]
  201. Pacheco, C.; Baião, A.; Ding, T.; Cui, W.; Sarmento, B. Recent Advances in Long-Acting Drug Delivery Systems for Anticancer Drug. Adv. Drug Deliv. Rev. 2023, 194, 114724. [Google Scholar] [CrossRef] [PubMed]
  202. Lukova, P.; Katsarov, P. Contemporary Aspects of Designing Marine Polysaccharide Microparticles as Drug Carriers for Biomedical Application. Pharmaceutics 2023, 15, 2126. [Google Scholar] [CrossRef] [PubMed]
  203. Lee, J.H.; Yeo, Y. Controlled drug release from pharmaceutical nanocarriers. Chem. Eng. Sci. 2015, 125, 75–84. [Google Scholar] [CrossRef]
  204. Desai, N.; Rana, D.; Salave, S.; Gupta, R.; Patel, P.; Karunakaran, B.; Sharma, A.; Giri, J.; Benival, D.; Kommineni, N. Chitosan: A Potential Biopolymer in Drug Delivery and Biomedical Applications. Pharmaceutics 2023, 15, 1313. [Google Scholar] [CrossRef] [PubMed]
  205. Jin, H.; Zhao, Z.; Lan, Q.; Zhou, H.; Mai, Z.; Wang, Y.; Ding, X.; Zhang, W.; Pi, J.; Evans, C.E.; et al. Nasal Delivery of Hesperidin/Chitosan Nanoparticles Suppresses Cytokine Storm Syndrome in a Mouse Model of Acute Lung Injury. Front. Pharmacol. 2021, 11, 592238. [Google Scholar] [CrossRef]
  206. Abdallah, H.M.; Abu Elella, M.H.; Abdel-Aziz, M.M. One-Pot Green Synthesis of Chitosan Biguanidine Nanoparticles for Targeting M. tuberculosis. Int. J. Biol. Macromol. 2023, 232, 123394. [Google Scholar] [CrossRef]
  207. Petkar, K.C.; Chavhan, S.; Kunda, N.; Saleem, I.; Somavarapu, S.; Taylor, K.M.G.; Sawant, K.K. Development of Novel Octanoyl Chitosan Nanoparticles for Improved Rifampicin Pulmonary Delivery: Optimization by Factorial Design. AAPS PharmSciTech 2018, 19, 1758–1772. [Google Scholar] [CrossRef]
  208. Niaz, T.; Sarkar, A.; Mackie, A.; Imran, M. Impact of Albumin Corona on Mucoadhesion and Antimicrobial Activity of Carvacrol Loaded Chitosan Nano-Delivery Systems under Simulated Gastro-Intestinal Conditions. Int. J. Biol. Macromol. 2021, 169, 171–182. [Google Scholar] [CrossRef]
  209. Yildiz-Peköz, A.; Akbal, O.; Tekarslan, S.H.; Sagirli, A.O.; Mulazimoglu, L.; Morina, D.; Cevher, E. Preparation and Characterization of Doripenem-Loaded Microparticles for Pulmonary Delivery. J. Aerosol Med. Pulm. Drug Deliv. 2018, 31, 347–357. [Google Scholar] [CrossRef]
  210. Hemmingsen, L.M.; Julin, K.; Ahsan, L.; Basnet, P.; Johannessen, M.; Škalko-Basnet, N. Chitosomes-In-Chitosan Hydrogel for Acute Skin Injuries: Prevention and Infection Control. Mar. Drugs 2021, 19, 269. [Google Scholar] [CrossRef] [PubMed]
  211. Abilova, G.K.; Kaldybekov, D.B.; Irmukhametova, G.S.; Kazybayeva, D.S.; Iskakbayeva, Z.A.; Kudaibergenov, S.E.; Khutoryanskiy, V.V. Chitosan/Poly(2-Ethyl-2-Oxazoline) Films with Ciprofloxacin for Application in Vaginal Drug Delivery. Materials 2020, 13, 1709. [Google Scholar] [CrossRef] [PubMed]
  212. Priya Dharshini, K.; Fang, H.; Ramya Devi, D.; Yang, J.-X.; Luo, R.-H.; Zheng, Y.-T.; Brzeziński, M.; Vedha Hari, B.N. pH-Sensitive Chitosan Nanoparticles Loaded with Dolutegravir as Milk and Food Admixture for Paediatric Anti-HIV Therapy. Carbohydr. Polym. 2021, 256, 117440. [Google Scholar] [CrossRef] [PubMed]
  213. Gondil, V.S.; Dube, T.; Panda, J.J.; Yennamalli, R.M.; Harjai, K.; Chhibber, S. Comprehensive Evaluation of Chitosan Nanoparticle Based Phage Lysin Delivery System; A Novel Approach to Counter S. pneumoniae Infections. Int. J. Pharm. 2020, 573, 118850. [Google Scholar] [CrossRef]
  214. Zareshahrabadi, Z.; Khorram, M.; Pakshir, K.; Tamaddon, A.-M.; Jafari, M.; Nouraei, H.; Ardekani, N.T.; Amirzadeh, N.; Irajie, C.; Barzegar, A.; et al. Magnetic Chitosan Nanoparticles Loaded with Amphotericin B: Synthesis, Properties and Potentiation of Antifungal Activity against Common Human Pathogenic Fungal Strains. Int. J. Biol. Macromol. 2022, 222, 1619–1631. [Google Scholar] [CrossRef]
  215. Pramanik, A.; Jones, S.; Gao, Y.; Sweet, C.; Begum, S.; Shukla, M.K.; Buchanan, J.P.; Moser, R.D.; Ray, P.C. A Bio-Conjugated Chitosan Wrapped CNT Based 3D Nanoporous Architecture for Separation and Inactivation of Rotavirus and Shigella Waterborne Pathogens. J. Mater. Chem. B 2017, 5, 9522–9531. [Google Scholar] [CrossRef]
  216. Liu, L.; Ma, Q.; Wang, S.; Gao, Y.; Zhu, C.; Zhao, W.; Sun, W.; Ma, H.; Sun, Y. Efficient Epidermal Delivery of Antibiotics by Self-Assembled Lecithin/Chitosan Nanoparticles for Enhanced Therapy on Epidermal Bacterial Infections. Int. J. Biol. Macromol. 2022, 218, 568–579. [Google Scholar] [CrossRef]
  217. Silva, M.; Calado, R.; Marto, J.; Bettencourt, A.; Almeida, A.; Gonçalves, L. Chitosan Nanoparticles as a Mucoadhesive Drug Delivery System for Ocular Administration. Mar. Drugs 2017, 15, 370. [Google Scholar] [CrossRef]
  218. Rashki, S.; Safardoust-Hojaghan, H.; Mirzaei, H.; Abdulsahib, W.K.; Mahdi, M.A.; Salavati-Niasari, M.; Khaledi, A.; Khorshidi, A.; Mousavi, S.G.A. Delivery LL37 by Chitosan Nanoparticles for Enhanced Antibacterial and Antibiofilm Efficacy. Carbohydr. Polym. 2022, 291, 119634. [Google Scholar] [CrossRef] [PubMed]
  219. Mosafer, J.; Sabbaghi, A.-H.; Badiee, A.; Dehghan, S.; Tafaghodi, M. Preparation, Characterization and In Vivo Evaluation of Alginate-Coated Chitosan and Trimethylchitosan Nanoparticles Loaded with PR8 Influenza Virus for Nasal Immunization. Asian J. Pharm. Sci. 2019, 14, 216–221. [Google Scholar] [CrossRef]
  220. Jearanaiwitayakul, T.; Sunintaboon, P.; Chawengkittikul, R.; Limthongkul, J.; Midoeng, P.; Chaisuwirat, P.; Warit, S.; Ubol, S. Whole Inactivated Dengue Virus-Loaded Trimethyl Chitosan Nanoparticle-Based Vaccine: Immunogenic Properties in Ex Vivo and in Vivo Models. Hum. Vaccines Immunother. 2021, 17, 2793–2807. [Google Scholar] [CrossRef]
  221. Acevedo-Villanueva, K.Y.; Lester, B.; Renu, S.; Han, Y.; Shanmugasundaram, R.; Gourapura, R.; Selvaraj, R. Efficacy of Chitosan-Based Nanoparticle Vaccine Administered to Broiler Birds Challenged with Salmonella. PLoS ONE 2020, 15, e0231998. [Google Scholar] [CrossRef] [PubMed]
  222. Helmy, Y.A.; Closs, G.; Jung, K.; Kathayat, D.; Vlasova, A.; Rajashekara, G. Effect of Probiotic E. coli Nissle 1917 Supplementation on the Growth Performance, Immune Responses, Intestinal Morphology, and Gut Microbes of Campylobacter Jejuni Infected Chickens. Infect. Immun. 2022, 90, e00337-22. [Google Scholar] [CrossRef]
  223. Bento, D.; Jesus, S.; Lebre, F.; Gonçalves, T.; Borges, O. Chitosan Plus Compound 48/80: Formulation and Preliminary Evaluation as a Hepatitis B Vaccine Adjuvant. Pharmaceutics 2019, 11, 72. [Google Scholar] [CrossRef] [PubMed]
  224. Zhao, K.; Li, S.; Li, W.; Yu, L.; Duan, X.; Han, J.; Wang, X.; Jin, Z. Quaternized Chitosan Nanoparticles Loaded with the Combined Attenuated Live Vaccine against Newcastle Disease and Infectious Bronchitis Elicit Immune Response in Chicken after Intranasal Administration. Drug Deliv. 2017, 24, 1574–1586. [Google Scholar] [CrossRef]
  225. Hajam, I.A.; Senevirathne, A.; Hewawaduge, C.; Kim, J.; Lee, J.H. Intranasally Administered Protein Coated Chitosan Nanoparticles Encapsulating Influenza H9N2 HA2 and M2e mRNA Molecules Elicit Protective Immunity against Avian Influenza Viruses in Chickens. Vet. Res. 2020, 51, 37. [Google Scholar] [CrossRef]
  226. Helmi, O.; Elshishiny, F.; Mamdouh, W. Targeted Doxorubicin Delivery and Release within Breast Cancer Environment Using PEGylated Chitosan Nanoparticles Labeled with Monoclonal Antibodies. Int. J. Biol. Macromol. 2021, 184, 325–338. [Google Scholar] [CrossRef]
  227. Naseer, F.; Ahmad, T.; Kousar, K.; Kakar, S.; Gul, R.; Anjum, S. Formulation of Surface-Functionalized Hyaluronic Acid-Coated Thiolated Chitosan Nano-Formulation for the Delivery of Vincristine in Prostate Cancer: A Multifunctional Targeted Drug Delivery Approach. J. Drug Deliv. Sci. Technol. 2022, 74, 103545. [Google Scholar] [CrossRef]
  228. Hu, H.; Qi, Q.; Dong, Z.; Yu, X.; Mo, Y.; Luo, J.; Wang, Y.; Du, S.; Lu, Y. Albumin Coated Trimethyl Chitosan-Based Targeting Delivery Platform for Photothermal/Chemo-Synergistic Cancer Therapy. Carbohydr. Polym. 2020, 241, 116335. [Google Scholar] [CrossRef]
  229. El-Dakroury, W.A.; Zewail, M.B.; Amin, M.M. Design, Optimization, and in-Vivo Performance of Glipizide-Loaded O-Carboxymethyl Chitosan Nanoparticles in Insulin Resistant/Type 2 Diabetic Rat Model. J. Drug Deliv. Sci. Technol. 2023, 79, 104040. [Google Scholar] [CrossRef]
  230. Wilson, B.; Mohamed Alobaid, B.N.; Geetha, K.M.; Jenita, J.L. Chitosan Nanoparticles to Enhance Nasal Absorption and Brain Targeting of Sitagliptin to Treat Alzheimer’s Disease. J. Drug Deliv. Sci. Technol. 2021, 61, 102176. [Google Scholar] [CrossRef]
  231. Li, B.; Wang, J.; Gui, Q.; Yang, H. Drug-Loaded Chitosan Film Prepared via Facile Solution Casting and Air-Drying of Plain Water-Based Chitosan Solution for Ocular Drug Delivery. Bioact. Mater. 2020, 5, 577–583. [Google Scholar] [CrossRef]
  232. Akhlaq, M.; Azad, A.K.; Fuloria, S.; Meenakshi, D.U.; Raza, S.; Safdar, M.; Nawaz, A.; Subramaniyan, V.; Sekar, M.; Sathasivam, K.V.; et al. Fabrication of Tizanidine Loaded Patches Using Flaxseed Oil and Coriander Oil as a Penetration Enhancer for Transdermal Delivery. Polymers 2021, 13, 4217. [Google Scholar] [CrossRef] [PubMed]
  233. Wei, H.; Liu, S.; Tong, Z.; Chen, T.; Yang, M.; Guo, Y.; Sun, H.; Wu, Y.; Chu, Y.; Fan, L. Hydrogel-Based Microneedles of Chitosan Derivatives for Drug Delivery. React. Funct. Polym. 2022, 172, 105200. [Google Scholar] [CrossRef]
  234. Pourjavadi, A.; Bagherifard, M.; Doroudian, M. Synthesis of Micelles Based on Chitosan Functionalized with Gold Nanorods as a Light Sensitive Drug Delivery Vehicle. Int. J. Biol. Macromol. 2020, 149, 809–818. [Google Scholar] [CrossRef]
  235. Chen, X.; Gu, J.; Sun, L.; Li, W.; Guo, L.; Gu, Z.; Wang, L.; Zhang, Y.; Zhang, W.; Han, B.; et al. Efficient Drug Delivery and Anticancer Effect of Micelles Based on Vitamin E Succinate and Chitosan Derivatives. Bioact. Mater. 2021, 6, 3025–3035. [Google Scholar] [CrossRef] [PubMed]
  236. Gu, H.; Chen, P.; Liu, X.; Lian, Y.; Xi, J.; Li, J.; Song, J.; Li, X. Trimethylated Chitosan-Coated Flexible Liposomes with Resveratrol for Topical Drug Delivery to Reduce Blue-Light-Induced Retinal Damage. Int. J. Biol. Macromol. 2023, 252, 126480. [Google Scholar] [CrossRef]
  237. Khodarahmi, M.; Abbasi, H.; Kouchak, M.; Mahdavinia, M.; Handali, S.; Rahbar, N. Nanoencapsulation of Aptamer-Functionalized 5-Fluorouracil Liposomes Using Alginate/Chitosan Complex as a Novel Targeting Strategy for Colon-Specific Drug Delivery. J. Drug Deliv. Sci. Technol. 2022, 71, 103299. [Google Scholar] [CrossRef]
  238. Ding, Y.; Yang, R.; Yu, W.; Hu, C.; Zhang, Z.; Liu, D.; An, Y.; Wang, X.; He, C.; Liu, P.; et al. Chitosan Oligosaccharide Decorated Liposomes Combined with TH302 for Photodynamic Therapy in Triple Negative Breast Cancer. J. Nanobiotechnol. 2021, 19, 147. [Google Scholar] [CrossRef] [PubMed]
  239. Jafernik, K.; Ładniak, A.; Blicharska, E.; Czarnek, K.; Ekiert, H.; Wiącek, A.E.; Szopa, A. Chitosan-Based Nanoparticles as Effective Drug Delivery Systems-A review. Molecules 2023, 28, 1963. [Google Scholar] [CrossRef] [PubMed]
  240. Aydin, R.S.T.; Pulat, M. 5-Fluorouracil encapsulated chitosan nanoparticles for pH-stimulated drug delivery: Evaluation of controlled release kinetics. J. Nanomater. 2012, 2012, 42. [Google Scholar] [CrossRef]
  241. Lim, E.K.; Huh, Y.M.; Yang, J.; Lee, K.; Suh, J.S.; Haam, S. pH-triggered drug-releasing magnetic nanoparticles for cancer therapy guided by molecular imaging by MRI. Adv. Mater. 2011, 23, 2436–2442. [Google Scholar] [CrossRef] [PubMed]
  242. Hu, X.; Wu, T.; Bao, Y.; Zhang, Z. Nanotechnology based therapeutic modality to boost anti-tumor immunity and collapse tumor defense. J. Control. Release 2017, 256, 26–45. [Google Scholar] [CrossRef]
  243. Liu, H.; Liu, W.; Tan, Z.; Zeng, Z.; Yang, H.; Luo, S.; Wang, L.; Xi, T.; Xing, Y. Promoting Immune Efficacy of the Oral Helicobacter pylori Vaccine by HP55/PBCA Nanoparticles against the Gastrointestinal Environment. Mol. Pharm. 2018, 15, 3177–3186. [Google Scholar] [CrossRef]
  244. Bhavsar, C.; Momin, M.; Gharat, S.; Omri, A. Functionalized and graft copolymers of chitosan and its pharmaceutical applications. Expert Opin. Drug Deliv. 2017, 14, 1189–1204. [Google Scholar] [CrossRef]
  245. Gierszewska, M.; Ostrowska-Czubenko, J.; Chrzanowska, E. pH-responsive chitosan/alginate polyelectrolyte complex membranes reinforced by tripolyphosphate. Eur. Polym. J. 2018, 101, 282–290. [Google Scholar] [CrossRef]
  246. Stasińska-Jakubas, M.; Hawrylak-Nowak, B. Protective, Biostimulating, and Eliciting Effects of Chitosan and Its Derivatives on Crop Plants. Molecules 2022, 27, 2801. [Google Scholar] [CrossRef]
  247. Riseh, R.S.; Hassanisaadi, M.; Vatankhah, M.; Babaki, S.A.; Barka, E.A. Chitosan as a Potential Natural Compound to Manage Plant Diseases. Int. J. Biol. Macromol. 2022, 220, 998–1009. [Google Scholar] [CrossRef]
  248. García-Carrasco, M.; Valdez-Baro, O.; Cabanillas-Bojórquez, L.A.; Bernal-Millán, M.J.; Rivera-Salas, M.M.; Gutiérrez-Grijalva, E.P.; Heredia, J.B. Potential Agricultural Uses of Micro/Nano Encapsulated Chitosan: A Review. Macromol 2023, 3, 614–635. [Google Scholar] [CrossRef]
  249. Doares, S.H.; Syrovets, T.; Weiler, E.W.; Ryan, C.A. Oligogalacturonides and Chitosan Activate Plant Defensive Genes through the Octadecanoid Pathway. Proc. Natl. Acad. Sci. USA 1995, 92, 4095–4098. [Google Scholar] [CrossRef] [PubMed]
  250. Sun, W.; Shahrajabian, M.H.; Petropoulos, S.A.; Shahrajabian, N. Developing Sustainable Agriculture Systems in Medicinal and Aromatic Plant Production by Using Chitosan and Chitin-Based Biostimulants. Plants 2023, 12, 2469. [Google Scholar] [CrossRef] [PubMed]
  251. Jogaiah, S.; Satapute, P.; De Britto, S.; Konappa, N.; Udayashankar, A.C. Exogenous Priming of Chitosan Induces Upregulation of Phytohormones and Resistance against Cucumber Powdery Mildew Disease Is Correlated with Localized Biosynthesis of Defense Enzymes. Int. J. Biol. Macromol. 2020, 162, 1825–1838. [Google Scholar] [CrossRef]
  252. Hidangmayum, A.; Dwivedi, P. Effect of Chitosan Seed Priming on Mungbean Seedlings Subjected to Different Levels of Water Potential. Acta Physiol. Plant. 2023, 45, 6. [Google Scholar] [CrossRef]
  253. Liu, H.; Zheng, Z.; Han, X.; Zhang, C.; Li, H.; Wu, M. Chitosan Soaking Improves Seed Germination of Platycodon Grandiflorus and Enhances Its Growth, Photosynthesis, Resistance, Yield, and Quality. Horticulturae 2022, 8, 943. [Google Scholar] [CrossRef]
  254. Razavizadeh, R.; Adabavazeh, F.; Komatsu, S. Chitosan Effects on the Elevation of Essential Oils and Antioxidant Activity of Carum copticum L. Seedlings and Callus Cultures under in Vitro Salt Stress. J. Plant Biochem. Biotechnol. 2020, 29, 473–483. [Google Scholar] [CrossRef]
  255. Ghule, M.R.; Ramteke, P.K.; Ramteke, S.D.; Kodre, P.S.; Langote, A.; Gaikwad, A.V.; Holkar, S.K.; Jambhekar, H. Impact of Chitosan Seed Treatment of Fenugreek for Management of Root Rot Disease Caused by Fusarium Solani under In Vitro and In Vivo Conditions. 3 Biotech 2021, 11, 290. [Google Scholar] [CrossRef]
  256. Nedved, E.L.; Kalatskaja, J.N.; Ovchinnikov, I.A.; Rybinskaya, E.I.; Kraskouski, A.N.; Nikalaichuk, V.V.; Hileuskaya, K.S.; Kulikouskaya, V.I.; Agabekov, V.E.; Laman, N.A. Growth Parameters and Antioxidant Activity in Cucumber Seedlings with the Application of Chitosan and Hydroxycinnamic Acids Conjugates under Salt Stress. Appl. Biochem. Microbiol. 2022, 58, 69–76. [Google Scholar] [CrossRef]
  257. Xu, D.; Li, H.; Lin, L.; Liao, M.; Deng, Q.; Wang, J.; Lv, X.; Deng, H.; Liang, D.; Xia, H. Effects of Carboxymethyl Chitosan on the Growth and Nutrient Uptake in Prunus Davidiana Seedlings. Physiol. Mol. Biol. Plants 2020, 26, 661–668. [Google Scholar] [CrossRef]
  258. Hoang, N.H.; Le Thanh, T.; Sangpueak, R.; Treekoon, J.; Saengchan, C.; Thepbandit, W.; Papathoti, N.K.; Kamkaew, A.; Buensanteai, N. Chitosan Nanoparticles-Based Ionic Gelation Method: A Promising Candidate for Plant Disease Management. Polymers 2022, 14, 662. [Google Scholar] [CrossRef] [PubMed]
  259. Narasimhamurthy, K.; Udayashankar, A.C.; De Britto, S.; Lavanya, S.N.; Abdelrahman, M.; Soumya, K.; Shetty, H.S.; Srinivas, C.; Jogaiah, S. Chitosan and Chitosan-Derived Nanoparticles Modulate Enhanced Immune Response in Tomato against Bacterial Wilt Disease. Int. J. Biol. Macromol. 2022, 220, 223–237. [Google Scholar] [CrossRef] [PubMed]
  260. Mondéjar-López, M.; Rubio-Moraga, A.; López-Jimenez, A.J.; García Martínez, J.C.; Ahrazem, O.; Gómez-Gómez, L.; Niza, E. Chitosan Nanoparticles Loaded with Garlic Essential Oil: A New Alternative to Tebuconazole as Seed Dressing Agent. Carbohydr. Polym. 2022, 277, 118815. [Google Scholar] [CrossRef] [PubMed]
  261. Gowda, S.; Sriram, S. Green Synthesis of Chitosan Silver Nanocomposites and Their Antifungal Activity against Colletotrichum Truncatum Causing Anthracnose in Chillies. Plant Nano Biol. 2023, 5, 100041. [Google Scholar] [CrossRef]
  262. Sathiyabama, M.; Manikandan, A. Foliar Application of Chitosan Nanoparticle Improves Yield, Mineral Content and Boost Innate Immunity in Finger Millet Plants. Carbohydr. Polym. 2021, 258, 117691. [Google Scholar] [CrossRef]
  263. Chouhan, D.; Mandal, P. Applications of Chitosan and Chitosan Based Metallic Nanoparticles in Agrosciences-A Review. Int. J. Biol. Macromol. 2021, 166, 1554–1569. [Google Scholar] [CrossRef]
  264. El Amerany, F.; Meddich, A.; Wahbi, S.; Porzel, A.; Taourirte, M.; Rhazi, M.; Hause, B. Foliar Application of Chitosan Increases Tomato Growth and Influences Mycorrhization and Expression of Endochitinase-Encoding Genes. Int. J. Mol. Sci. 2020, 21, 535. [Google Scholar] [CrossRef]
  265. Hao, T.; Yang, Z.; Liang, J.; Yu, J.; Liu, J. Foliar Application of Carnosine and Chitosan Improving Drought Tolerance in Bermudagrass. Agronomy 2023, 13, 442. [Google Scholar] [CrossRef]
  266. Hawrylak-Nowak, B.; Dresler, S.; Rubinowska, K.; Matraszek-Gawron, R. Eliciting Effect of Foliar Application of Chitosan Lactate on the Phytochemical Properties of Ocimum basilicum L. and Melissa officinalis L. Food Chem. 2021, 342, 128358. [Google Scholar] [CrossRef] [PubMed]
  267. Sheikhalipour, M.; Esmaielpour, B.; Behnamian, M.; Gohari, G.; Giglou, M.T.; Vachova, P.; Rastogi, A.; Brestic, M.; Skalicky, M. Chitosan–Selenium Nanoparticle (Cs–Se NP) Foliar Spray Alleviates Salt Stress in Bitter Melon. Nanomaterials 2021, 11, 684. [Google Scholar] [CrossRef]
  268. Ji, H.; Wang, J.; Chen, F.; Fan, N.; Wang, X.; Xiao, Z.; Wang, Z. Meta-Analysis of Chitosan-Mediated Effects on Plant Defense against Oxidative Stress. Sci. Total Environ. 2022, 851, 158212. [Google Scholar] [CrossRef]
  269. Zhang, Z.; Jin, F.; Wu, Z.; Jin, J.; Li, F.; Wang, Y.; Wang, Z.; Tang, S.; Wu, C.; Wang, Y. O-acylation of chitosan nanofibers by short-chain and long-chain fatty acids. Carbohydr. Polym. 2017, 177, 203–209. [Google Scholar] [CrossRef] [PubMed]
  270. Wedmore, I.; McManus, J.G.; Pusateri, A.E.; Holcomb, J.B. A special report on the chitosan-based hemostatic dressing: Experience in current combat operations. J. Trauma Acute Care Surg. 2006, 60, 655–658. [Google Scholar] [CrossRef] [PubMed]
  271. Malmquist, J.P.; Clemens, S.C.; Oien, H.J.; Wilson, S.L. Hemostasis of oral surgery wounds with the HemCon Dental Dressing. J. Oral Maxillofac. Surg. 2008, 66, 1177–1183. [Google Scholar] [CrossRef] [PubMed]
Scheme 1. Synthetic strategy for the preparation of CS.
Scheme 1. Synthetic strategy for the preparation of CS.
Molecules 28 07659 sch001
Scheme 2. Synthesis scheme of chitosan derivatives through alkylation and acylation processes.
Scheme 2. Synthesis scheme of chitosan derivatives through alkylation and acylation processes.
Molecules 28 07659 sch002
Scheme 3. Synthesis of chitosan derivatives bearing Schiff bases.
Scheme 3. Synthesis of chitosan derivatives bearing Schiff bases.
Molecules 28 07659 sch003
Scheme 4. Synthesis of quaternary ammonium salt, N-trimethyl chitosan derivatives. The red color represents the functional group of quaternary ammonium salt.
Scheme 4. Synthesis of quaternary ammonium salt, N-trimethyl chitosan derivatives. The red color represents the functional group of quaternary ammonium salt.
Molecules 28 07659 sch004
Scheme 5. Synthesis of chitosan quaternary ammonium derivatives through a quaternary ammonium source. The red color represents the functional group of quaternary ammonium salt.
Scheme 5. Synthesis of chitosan quaternary ammonium derivatives through a quaternary ammonium source. The red color represents the functional group of quaternary ammonium salt.
Molecules 28 07659 sch005
Scheme 6. Synthesis of chitosan derivatives bearing pyridinium salt. The red color represents the functional group of pyridinium salt.
Scheme 6. Synthesis of chitosan derivatives bearing pyridinium salt. The red color represents the functional group of pyridinium salt.
Molecules 28 07659 sch006
Scheme 7. Synthesis of quaternary ammonium chitosan derivatives followed by click chemistry. The red color represents the functional group of quaternary ammonium salt.
Scheme 7. Synthesis of quaternary ammonium chitosan derivatives followed by click chemistry. The red color represents the functional group of quaternary ammonium salt.
Molecules 28 07659 sch007
Scheme 8. Synthetic strategy for guanidine chitosan derivatives. The red color represents the functional group of guanidine.
Scheme 8. Synthetic strategy for guanidine chitosan derivatives. The red color represents the functional group of guanidine.
Molecules 28 07659 sch008
Scheme 9. Synthetic strategy of guanidine chitosan salt derivatives. The red color represents the functional group of guanidine.
Scheme 9. Synthetic strategy of guanidine chitosan salt derivatives. The red color represents the functional group of guanidine.
Molecules 28 07659 sch009
Scheme 10. Synthetic strategy of biguanidine/polyaniline composite. The red color represents the functional group of guanidine.
Scheme 10. Synthetic strategy of biguanidine/polyaniline composite. The red color represents the functional group of guanidine.
Molecules 28 07659 sch010
Scheme 11. Synthetic strategy of heterocyclic chitosan derivatives through click chemistry. The red color represents the newly constructed triazole moiety.
Scheme 11. Synthetic strategy of heterocyclic chitosan derivatives through click chemistry. The red color represents the newly constructed triazole moiety.
Molecules 28 07659 sch011
Scheme 12. Synthetic strategy of furan-modified chitosan derivatives through Diels–Alder click reaction. The red color represents the creation of new bonds through Diels–Alder click reaction.
Scheme 12. Synthetic strategy of furan-modified chitosan derivatives through Diels–Alder click reaction. The red color represents the creation of new bonds through Diels–Alder click reaction.
Molecules 28 07659 sch012
Scheme 13. Synthetic strategy of heterocyclic chitosan derivatives through N-functionalized reaction. The red color represents the addition of new heterocyclic moiety through direct N-functionalization reaction.
Scheme 13. Synthetic strategy of heterocyclic chitosan derivatives through N-functionalized reaction. The red color represents the addition of new heterocyclic moiety through direct N-functionalization reaction.
Molecules 28 07659 sch013
Figure 1. Mechanism of antimicrobial effects of chitosan and chitosan-based materials against Gram-positive and Gram-negative bacteria (created with BioRender.com, accessed on 27 September 2023).
Figure 1. Mechanism of antimicrobial effects of chitosan and chitosan-based materials against Gram-positive and Gram-negative bacteria (created with BioRender.com, accessed on 27 September 2023).
Molecules 28 07659 g001
Figure 2. The mechanisms of release of drugs/antigens from chitosan nanoparticles (created with Microsoft Power Point 2010 and Chemdraw 19.1).
Figure 2. The mechanisms of release of drugs/antigens from chitosan nanoparticles (created with Microsoft Power Point 2010 and Chemdraw 19.1).
Molecules 28 07659 g002
Figure 3. Schematic representation of the application of chitosan-based nanoparticles in plant agriculture (created with Microsoft PowerPoint 2010, Chemdraw 19.1, and BioRender.com, accessed on 27 September 2023).
Figure 3. Schematic representation of the application of chitosan-based nanoparticles in plant agriculture (created with Microsoft PowerPoint 2010, Chemdraw 19.1, and BioRender.com, accessed on 27 September 2023).
Molecules 28 07659 g003
Table 1. Patents on chitosan-based materials in antimicrobial, drug delivery, and agricultural applications.
Table 1. Patents on chitosan-based materials in antimicrobial, drug delivery, and agricultural applications.
Patent AreaApplicationFinal Form References
Biomedical/AntimicrobialAs a teat sealant wherein CS hydrogels accelerate involution and prevent infection of mammary glandHydrogels[33]
Biomedical/AntimicrobialAn adsorbent biodegradable wound dressingFilms[34]
Biomedical/AntimicrobialCS coating in metallic nanoparticles as an anti-biofilm agent on medically implantable articlesNanoparticles/powder[35]
Biomedical/AntimicrobialFor coating Listeria iuanuii, to enhance negative surface charge and thus immune responseCS oligosaccharide[36]
Biomedical/AntimicrobialAntimicrobial wound dressing material with quaternized CSQuaternized CS sponges[37]
Biomedical/AntimicrobialFormulation of CS with metallic nanoparticles for treating skin lesions/infectionsGel matrix[38]
Biomedical/Drug deliveryCS hydrogel used for targeted drug deliveryHydrogels[39]
Biomedical/Drug deliveryLow-toxicity and high-affinity CS derivative for drug deliveryQuaternized, acylated CS derivative[40]
Biomedical/Drug deliveryDelivery of IL17RC protein that function by improving nasal adsorptionNanoparticles[41]
Biomedical/Drug deliveryCS–nucleic acid polyplexes for delivery of nucleic acid encoding IL-2Nanoparticles[42]
Biomedical/Drug deliveryTreatment of transected peripheral nerve injuriesCS hydrogel[43]
AgriculturalGel microspheres of CS with manganese and prothioconazole for use against pestsGel microspheres[44]
AgriculturalAntifungal activity in plantsCS derivative[45]
AgriculturalPesticides that comprise CS and berberinePowder, suspension[46]
AgriculturalLiquid fertilizer with CS oligosaccharides and amino acidsLiquid[47]
Table 3. Applications of CS-based nano/micro particles as drug/vaccine carriers for infectious pathogens.
Table 3. Applications of CS-based nano/micro particles as drug/vaccine carriers for infectious pathogens.
CS
Derivative/Nanomaterials
PreparationInterferenceReferences
Hesperidin-loaded CS nanoparticleEmulsification and evaporation methods
  • Nasal delivery of the anti-inflammatory HPD compound to inflammatory lungs;
  • Enhance cellular uptake in the inflammatory microenvironment compared with free HPD;
  • Suppress the cytokine storm syndrome and acute lung injury/acute respiratory distress syndrome in a murine model of inflammatory lung disease.
[205]
CS biguanidine nanoparticleOne pot, green, ionic gelation method
  • Inhibitory activity against the growth of three different Mycobacterium tuberculosis pathogens (sensitive, MDR, and XDR) compared with isoniazid drug (standard anti-tuberculosis drug).
[206]
Rifampicin-loaded octanoyl CS nanoparticleDouble emulsion
solvent evaporation
technique
  • Pulmonary delivery;
  • Optimization of drug release, solubility, and pulmonary release of rifampicin through this carrier;
  • Aim at improving the interventions for tuberculosis.
[207]
Carvacrol-loaded bovine serum albumin (BSA) CS nanoparticleIonic gelation method
  • Mucosal delivery;
  • Ensure delayed release and limited degradation in the gastric conditions;
  • Effective eradication of Salmonella enterica serovar Typhi.
[208]
Doripenem-loaded CS
micro-particles
Ionic gelation, spray-drying method
  • Effective pulmonary delivery of doripenem;
  • Significant effect on reducing the infection by P. aeruginosa.
[209]
Chlorhexidine-infused CS One-pot method
  • Epidermal delivery;
  • Show improved antibacterial activity against S. aureus and S. epidermidis.
[210]
Ciprofloxacin-loaded
CS/poly
(2-ethyl-2-oxazoline)
Solvent evaporation method
  • Antimicrobial properties against S. aureus and E. coli;
  • Mucoadhesive vaginal drug delivery system for sheep.
[211]
CS nanoparticle loaded with
dolutegravir
Spray-drying method
  • High drug loading and releasing activity;
  • Anti-HIV activity.
[212]
CS nanoparticle loaded with phage endolysin Cpl-1Ionic gelation method
  • Mucoadhesive drug delivery;
  • Increased therapeutic applicability of endolysins.
[213]
Magnetic CS loaded with
amphotericin B
Chemical coprecipitation method followed by surface coating
  • Stable and controlled delivery of amphotericin B;
  • Interfere with biofilm formation and fungicidal effect.
[214]
CS-wrapped carbon nano tubes Cross-linking method of CS and CNTs
  • Separation, identification, and eradication of rotavirus and Shigella, waterborne pathogens in water samples of diverse sources.
[215]
CS/lecithin nanoparticles loaded with antibioticsCentral rotatable composite design method
  • Epidermal delivery;
  • Increase the accumulation of antibiotics in the epidermis with higher retention ratio compared to the commercial formulations;
  • Test against E. coli and S. aureus.
[216]
CS–hyaluronic acid-based nanoparticle containing ceftazidimeIonic gelation method
  • Mucoadhesive drug delivery;
  • Increase interaction with ocular surface and drug residence time;
  • Prolong drug release profile.
[217]
CS nanoparticle containing LL-37Ionotropic gelation method
  • Increase half-life and prolonged LL37 biological activity;
  • Inhibition of high biofilm formation by Methicillin-resistant S. aureus when compared to LL37 alone.
[218]
Inactivated PR8 influenza virus-loaded CS alginate and trimethyl CS nanoparticlesIonic gelation method
  • Immunoadjuvant potential after nasal immunization;
  • Exhibited higher IgG2a/IgG1 ratio as criteria for Th1-type immune response;
  • Efficient intranasal antigen delivery system for nasal vaccines.
[219]
Inactivated Dengue virus (DENV)-loaded trimethyl CS nanoparticleIonotropic gelation method
  • Highly immunogenic, inducing greater levels of antibodies (total IgG, IgG1, IgG2a and neutralizing antibodies) and T cells (activated CD4⁺ and CD8⁺ T cells) against DENV-2 compared to soluble DENV-2 immunogens.
[220]
CS-based nanoparticle vaccine loaded with Salmonella Enteritidis outer membrane proteins and flagellin proteins Ionic gelation method
  • Mucosal delivery;
  • Induce specific immune responses against the bacteria and reduced cecal colonization in birds;
  • Almost 65.9% birds which were vaccinated had cleared the infection.
[221]
E. coli Nissle 1917 microencapsulated
CS–alginate
Ionic gelation method
  • Reduce Campylobacter jejuni colonization in infected chickens;
  • Improve the intestinal morphology, increase immune responses of treated chickens via likely activation of the Th1, Th2, and Th17 pathways.
[222]
Mast cell activator C48/80 with CS nanoparticleMixing of C48/80 compound with CS, lyophilization
  • Exhibit adsorption of model antigen C48/80 and biocompatible onto the nanoparticle;
  • Demonstrate first time for the hepatitis B surface antigen loaded Chi-C48/80 nanoparticle as nasal vaccination strategy.
[223]
Newcastle disease virus (NDV/La Sota) and infectious bronchitis virus (IBV/H120)-loaded quaternized CS (-2-hydroxypropyl trimethyl ammonium chloride CS (N-2-HACC) and N, O-carboxymethyl CS) nanoparticlesPolyelectrolyte composite method
  • Newcastle disease and Infectious Bronchitis elicit immune response in chickens after intranasal administration;
  • Inducement of higher titers of IgG and IgA antibodies;
  • Increase the proliferation of lymphocytes and induce higher levels of cytokines, including IL-2, IL-4, and IFN- γ .
[224]
CS nanoparticles encapsulating influenza H9N2 HA2 and M2e mRNA moleculesAnionic gelation method
  • Delivery of immunogenic antigens to antigen presenting cells;
  • Effective penetration of the mucosal barrier to reach immune initiation sites;
  • Efficient release of mRNA molecules into antigen presenting cells;
  • Higher systemic IgG, mucosal IgA antibody response.
[225]
Note: HPD: hesperidin, MDR: multidrug-resistant, XDR: extensively drug-resistant, P. aeruginosa: Pseudomonas aeruginosa, S. aureus: Staphylococcus aureus, S. epidermidis: Staphylococcus epidermidis, E. coli: Escherichia coli.
Table 4. Application of chitosan and its functionalized nanoparticles for drug delivery system.
Table 4. Application of chitosan and its functionalized nanoparticles for drug delivery system.
FormDrug Carrier SystemDrug or EnzymesPreparationInterferenceReferences
NanoparticleAnti-hMAM and anti-HER2 PEGylated CS nanoparticleDoxorubicin (DOX)Mixing the DOX in the PEG-CS solution, cross-linking with TPP.
  • Cytotoxic against MCF-7 cancer cells (breast cancer).
[226]
Hyaluronic acid-coated thiolated CS nano
particle
VincristineIonic gelation process
  • In vitro anticancer activity against cancerous prostate cells.
[227]
Human serum albumin (HSA)-coated trimethyl CSIR780 (I) or bufalin (B)Ionic gelation process and electrostatic absorption
  • Inhibition of cell proliferation and mitochondrion activity of metastatic 4T1 breast cancer cells;
  • Enhance the tumor-targeting and penetrating capability for photo thermal therapy, efficient prevention of lung metastasis of breast cancer.
[228]
O-carboxymethyl CS
nanoparticle
GlipizideIonotropic gelation method
  • Significant impact in insulin-resistant/type 2 diabetes.
[229]
CS
nanoparticle
SitagliptinIonic gelation method
  • Increase symptomatic relief of Alzheimer’s disease by increasing the level of sitagliptin in the brain.
[230]
FilmCS-based filmBrimonidine tartrate (BT)Dissolution method
  • High corneal permeability with fast drug release for potential ocular drug delivery.
[231]
PatchesCS, thiolated CS patches with flaxseed oil and coriander oilTizanidineThe solvent casting method
  • In vitro transdermal drug release and drug permeation, skin irritation;
  • In vivo application, pharmacokinetics analysis, and stability studies.
[232]
HydrogelCarboxymethyl CS–silk
fibroin
peptide/oxidized pullulan (CMCS-SFP/OPL) hydrogel-based microneedle
Salvia
miltiorrhiza
Polydimethylsiloxane (PDMS) mold
  • Direct drug release from HFM-1;
  • Alternative transdermal delivery of water-soluble Salvia miltiorrhiza.
[233]
MicellesCS thiourea with gold nanorodsPaclitaxelRing opening polymerization, dialysis,
gold-thiolate complex
formation
  • Light-sensitive drug delivery vehicle;
  • Cytotoxicity against MCF7 cells.
[234]
O-CMCTS-VES micelles
nanoparticles
DoxorubicinDehydrative condensation of the carboxyl group of the carboxymethyl CS and the amino group of the vitamin E succinate, self-assembled nano-micelles
  • Efficient cellular uptake by HePG2 cancer cells (human liver cancer) and inhibition of tumor (62.7%).
[235]
LiposomeTrimethylated CS-coated
flexible liposomes
ResveratrolSolvent injection method,
electrostatic adsorption
  • Topical drug (Resveratrol) delivery to reduce blue-light-induced retinal damage;
  • Modulation for the mitochondrial membrane potential and protection of ARPE-19 cells from damage by H2O2.
[236]
Aptamer-functionalized liposomal coated with calcium alginate/CS/PEC5-
Fluorouracil (AFL5-FU)
Optimized thin film method, freeze–thaw process
  • Oral formulation for colon cancer therapy;
  • Cytotoxicity against on the colon cancer cell line, HT-29 (IC50; 66.53 3M).
[237]
CS oligosaccharide (CO)
nanoparticle based on liposomes
Photosensitizer HPPH and the
hypoxia-activated prodrug TH302
(CO-HPPH-
TH302/Lipo)
O/W emulsification method,
mixing the
lipids, drugs, and CO-OA
  • Activation for PDT and chemotherapy by HPPH and TH302;
  • Inhibition of the growth of CD44-overexpressing tumors with minimal damage to normal tissues by CD44+-targeted liposomes.
[238]
Note: Anti-hMAM: anti-human mammaglobin, Anti-HER2: anti-human epidermal growth factor, HFM-1: Helicase for Meiosis 1, O-CMCTS-VES: carboxymethyl CS/vitamin E succinate nano-micellar System, PEC: photoelectrolyte complexes, HPPH: 2-[1-hexyloxyethyl]-2-devinyl pyropheophorbide-a (Photochlor), TH302: Evofosfamide, CO: CS oligosaccharide, OA: oleic acid, PDT: photodynamic therapy.
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

Shrestha, R.; Thenissery, A.; Khupse, R.; Rajashekara, G. Strategies for the Preparation of Chitosan Derivatives for Antimicrobial, Drug Delivery, and Agricultural Applications: A Review. Molecules 2023, 28, 7659. https://doi.org/10.3390/molecules28227659

AMA Style

Shrestha R, Thenissery A, Khupse R, Rajashekara G. Strategies for the Preparation of Chitosan Derivatives for Antimicrobial, Drug Delivery, and Agricultural Applications: A Review. Molecules. 2023; 28(22):7659. https://doi.org/10.3390/molecules28227659

Chicago/Turabian Style

Shrestha, Rajeev, Anusree Thenissery, Rahul Khupse, and Gireesh Rajashekara. 2023. "Strategies for the Preparation of Chitosan Derivatives for Antimicrobial, Drug Delivery, and Agricultural Applications: A Review" Molecules 28, no. 22: 7659. https://doi.org/10.3390/molecules28227659

Article Metrics

Back to TopTop