Next Article in Journal
PKCeta Promotes Stress-Induced Autophagy and Senescence in Breast Cancer Cells, Presenting a Target for Therapy
Next Article in Special Issue
Immunogenic Cell Death Enhances Immunotherapy of Diffuse Intrinsic Pontine Glioma: From Preclinical to Clinical Studies
Previous Article in Journal
Efficient Delivery of DNA Using Lipid Nanoparticles
Previous Article in Special Issue
Cancer Immunotherapy and Delivery System: An Update
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Herb Polysaccharide-Based Drug Delivery System: Fabrication, Properties, and Applications for Immunotherapy

1
School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
2
Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan Province, Kunming 650500, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to the work.
Pharmaceutics 2022, 14(8), 1703; https://doi.org/10.3390/pharmaceutics14081703
Submission received: 21 June 2022 / Revised: 7 August 2022 / Accepted: 10 August 2022 / Published: 15 August 2022
(This article belongs to the Special Issue Drug Repurposing and Delivery Systems for Immunotherapy)

Abstract

:
Herb polysaccharides (HPS) have been studied extensively for their healthcare applications. Though the toxicity was not fully clarified, HPS were widely accepted for their biodegradability and biocompatibility. In addition, as carbohydrate polymers with a unique chemical composition, molecular weight, and functional group profile, HPS can be conjugated, cross-linked, and functionally modified. Thus, they are great candidates for the fabrication of drug delivery systems (DDS). HPS-based DDS (HPS-DDS) can bypass phagocytosis by the reticuloendothelial system, prevent the degradation of biomolecules, and increase the bioavailability of small molecules, thus exerting therapeutic effects. In this review, we focus on the application of HPS as components of immunoregulatory DDS. We summarize the principles governing the fabrication of HPS-DDS, including nanoparticles, micelles, liposomes, microemulsions, hydrogels, and microneedles. In addition, we discuss the role of HPS in DDS for immunotherapy. This comprehensive review provides valuable insights that could guide the design of effective HPS-DDS.

1. Introduction

Polysaccharides, which are natural high-molecular-weight compounds, are important biopolymers. They are composed of monosaccharide subunits joined by glycosidic linkages [1]. Typically, polysaccharides contain more than 10 monosaccharides, and their molecular weight (Mw) ranges from thousands to millions Da [2]. So far, more than 300 kinds of natural polysaccharides have been identified from fungi, plants, animals, algae, and bacteria [3]. Hence, they represent a fundamental renewable resource of biomass that can replace fossil-based polymeric materials in commodities and industrial applications [4]. Compared with synthetic materials, polysaccharides have unique biological properties, include hydrophilicity [5], good stability [6], abundant availability [7], bioadhesion [8], and low toxicity. Moreover, they often show low immunogenicity [9], biodegradability [10], and ease of chemical modification [11]. Polysaccharides remain popular candidates for fabricating biomaterials for drug delivery systems (DDS). For example, hyaluronic acid (HA), which can form gels, is used to produce scaffolds with very intriguing mechanical properties [12]. Chitosan (CS) [13], known for its anti-bacterial and film-forming properties, is widely used in advanced nanotechnology. Alginate is a natural polysaccharide that can easily be chemically modified or combined with other components to produce various functionalities. Furthermore, alginate derivatives are appealing not only because they can be used in biomedicine but also because of their potential in bioelectronics, as they can act as the interface between human tissues and electronic devices or even serve as electronic components themselves [14].
In addition to beneficial material properties, some polysaccharides also have significant biological activities [15]. In particular, polysaccharides derived from herbs, such as various Chinese herb medicines, have attracted attention for their immunostimulatory activities, although the detailed mechanism of their effects is currently under exploration [16]. Numerous studies indicate that HPS can be used in many fields and have diverse therapeutic properties and immunostimulatory activities [17], including antioxidation [18], antitumor [19], and wound-healing effects [20]. For example, Panax ginseng polysaccharides can regulate the immune system and induce anti-cancer effects via anti-complementary activity, peritoneal macrophage activation, and cytotoxicity against natural killer (NK) cells [21]. Astragalus is a tonic herb widely used to improve the immune status of the body. According to reports, polysaccharides from Astragalus can regulate the activities of immune organs and immune cells and promote the release of immune mediators (such as IL-2, IL-6, TNF-α, and IFN-γ) [22]. Dendrobium officinale polysaccharides can remarkably inhibit tumor growth by promoting macrophage polarization from the M2 phenotype to the M1 phenotype in the tumor microenvironment (TME) [23]. Bletilla striata polysaccharides are used to prepare composite aerogels that exhibit strong antibacterial and hemostatic activity, reducing inflammation and rapidly enhancing wound re-epithelialization by down-regulating TNF-α and up-regulating VEGF-A. These results prove that HPS possess excellent immunoregulatory bioactivities [24].
The combination of potential pharmacological activity and useful (bio)physical properties increases the value of HPS as building blocks for DDS, which are active treatment agents. Previous studies have reported the synthesis of HPS-DDS, such as nanoparticles (NPs) [25,26], micelles [27], liposomes [28], microemulsions [29], hydrogels [30], and microneedles [31], for the treatment of various diseases. These studies show that HPS can serve as drug carriers [32], stabilizers [33], solubilizers [34], and emulsifiers [35]. Considering their bioactivity, these HPS can also act as adjuvants [36], immunomodulators [37], and bioactive macromolecular drugs [38] that exert synergistic effects or amplify the therapeutic activities of primary drugs.
Several reviews have focused on DDS based on natural polysaccharides such as CS [39], HA [40], dextran [41], and alginate [42] and highlighted their material properties. However, the use of HPS for DDS with immunomodulatory activity has rarely been reviewed. In addition, the application of HPS in DDS have rarely been reviewed. Accordingly, this review discusses the main essential properties of HPS-based DDS, including their chemical properties and immunostimulatory mechanisms. In addition, the principle of HPS-DDS fabrication and the role of HPS in DDS are summarized. Finally, this review discusses the recent progress in the applications of HPS-DDS and addresses the prospects for future studies. The aim of this review is to offer a comprehensive understanding of the current research and development of HPS-DDS and provide a framework for the fabrication of novel HPS-DDS in the future.

2. Immunomodulatory Activity of HPS

More than 1000 kinds of HPS from herb plants have been demonstrated to have immune-related activities [43,44]. Table 1 shows the effect of various HPS (e.g., Bletilla striata, Angelica sinensis, and Astragalus membranaceus) on immune-related diseases. Notably, polysaccharides with immunomodulatory activities can directly or indirectly interact with the immune system [45]. In general, HPS exert immune effects by regulating immune cells (macrophages, dendritic cells (DCs), T cells, NK cells, etc.) [46] and promoting cell–cell communication [47]. Once a pathogen enters the immune system, it is recognized and processed by antigen-presenting cells (APC). These cells present antigens to T cells and induce T cell differentiation, leading to a series of immune responses [48]. For example, after microneedle administration, DCs prime T cells, thereby playing an immune role [49]. In TME, macrophages activate T cells, and T cells can also be affected directly [50,51]. According to the literature, traditional HPS such as honeysuckle polysaccharides [52], tea polysaccharides [53], and lychee polysaccharides [54] can promote the development of immune organs, participate in the body’s immune response and defense, and thus maintain an immune balance.
HPS perform immunomodulation through multiple modes and targets in order to regulate immune function, thereby improving biological immunity at the organ, tissue, cellular, and molecular levels [55,56]. Studies have shown that most HPS perform immunomodulation by binding to cell surface receptors, stimulating linked signaling channels, and promoting cytokine expression [45]. At present, the key HPS receptors identified include TLR-4 [57], scavenger receptor [58], mannose receptors (MRs) [59], MAPK [60], CR3/CD14 [61], and Dectin-1 [62]. In addition, other receptors and signaling channels can also mediate the immunomodulatory effects of HPS [45]. One example of the typical receptor-mediated immunomodulatory mechanisms underlying the effects of HPS includes the action of Angelica sinensis polysaccharides (ASPs) on renal leukocytes. These polysaccharides act on TLR-2 and its downstream MAPK and NF-κB signaling pathways as well as on molecules downstream of TLR4, such as MAPK and NF-κB. Accordingly, they upregulate MHC-II, TNF-α, and IL-8 [63]. Astragalus polysaccharides (APSs) act on dendritic cells (DCs) via the JAK/STAT signaling pathway [64]. Hibiscus sabdariffa Linn. polysaccharides act on RAW264.7 cells by activating the MAPK and NF-κB signal pathway, phosphorylating ERK, JNK, p38, and p65, and promoting the expression of iNOS, IL-6, IL-1α, TNF-α, and IL-6 [65]. Together, the findings suggest that the immunomodulatory effects of HPS do not follow a single pattern. Instead, these polysaccharides regulate the body’s immune function via multiple pathways and targets, thereby boosting biological immunity. It is worth noting that the immunomodulatory effects of HPS on different immune cells involve different receptors and signaling pathways, and these polysaccharides also exert regulatory effects on intercellular messenger molecules.
Numerous studies have shown that molecular weight [66], monosaccharide composition and ratio [67], glycoside bond type [68], and grading structure [69] can influence the immune activity of HPS. An excessively high molecular weight can reduce the exposure of the active sites on HPS, thus limiting their bioactivity. However, if the molecular weight is too low, the number of active functional groups becomes insufficient, leading to poor bioactivity [70,71,72]. The monosaccharide composition also plays an important role in the immune activity of HPS. Glucose, galactose, and mannose are more easily recognized by receptors on the cell surface and trigger a series of immune responses via macrophages [45]. Glucuronic acid can enhance the immune activity of polysaccharides [73]. The β-(1→3) glycosidic bond is a non-reducing glycosidic bond, increasing the stability and tolerance of polysaccharides and reducing its effect on polysaccharides activity [74,75,76].
The charge and the abundant active groups on the surface of HPS can also affect its immune activity. Research shows that the negatively charged HPS can avoid the attack of macrophages by electrostatic attraction, which can improve the efficacy of the immunomodulatory activity [77]. That is because a large number of negatively charged polyanionic HPS are more likely to interact with basic proteins in the body, recognize and control the activity of immune cells, reduce immunogenic response, and thus regulate their immune activity [78]. In addition, Wanwan Xiao et al. [79] showed that the four types of Platycodon grandiflorum Polysaccharides show different immune activity, and the higher uronic acid content exhibited better immune-stimulating activity. Due to the carboxyl groups being abundant in acidic polysaccharides, it has the ability to provide hydrogen to pathogenic sites for reducing inflammation [80]. Moreover, the expression of uronic acid receptor on of tumor tissues and immune cells is higher, which is conducive to the accumulation of HPS [81].
Table 1. Summary of herb polysaccharides and their bioactivities.
Table 1. Summary of herb polysaccharides and their bioactivities.
Herb
Polysaccharides
Molecular WeightMonosaccharide Composition and Proportion of MonosaccharideMain Types of Glycosidic BondsPharmacological
Effect
References
Bletilla striata polysaccharides8.354 × 104 Da
1.26 × 104 Da
Man and Glc in the ratio of 4.0:1.0
Man and Glc in the ratio of 3.0:1.0
Backbone composed of β-1,4-linked Manp
Backbone composed of β-1,4-linked Glcp
Immunomodulatory[76]
1.46 × 105 DaMan and Glc in the ratio of 2.4:1.0 α-Man and β-Glc residuesAnti-inflammatory[82]
Mw:3.73 × 105 g/mol
Mn:6.75 × 104 g/mol
Man and Glc in the ratio of 2.946:1Composed of 1,4-linked Glcp
Branches composed of 1,3-linked Manp and
1,3-linked Glcp
Anti-inflammatory[83]
2.35 × 105 DaMan, Glc, and Gal in the ratio of 9:2.6:1.0Composed of 1,4-linked Manp and 1,4-linked Glcp in a molar ratio of 2:1Immunomodulatory [84]
Angelica sinensis polysaccharides1.0 × 105 DaGlcα-(1→6)-glucanImmunomodulatory[85]
44,000 Daarabinose and galactose, as well as xylose, rhamnose and mannose1,5 linked, galactose containing 1,4 and 1,4,6 linked units, xylose 1,4 linked, rhamnose 1,2 linked, and terminal mannose [86]
American Ginseng polysaccharides Arabinose, rhamnose, mannose, glucose, and glucose acid Immunomodulatory [87]
3.1 kDaGlucose (Glc) and galactose (Gal) in a molar ratio of 1:1.15 Anti-inflammatory[88]
Astragalus membranaceus
polysaccharides
Glucose, in addition to rhamnose, galactose, arabinose, xylose, mannose, glucuronic acid, and galacturonic acid Immunoregulatory, antitumor, anti-inflammatory, and antiviral [89]
Lycium barbarum L. polysaccharides4.2 × 104 Da
4.1 × 104 Da
Glucose, mannose, and galactose
Glc:Man:Gal = 1:0.4:0.1
(1→3)-linked α-d-glucopyranosyl residues
(1→2)-linked α-d-mannopyranosyl residues
a-d-(1,3)-Glcp
a-d-(1,2)-Manp-p-d-Galp
(1→2)-linked α-d-mannopyranosyl residues, β-D-galactopyranosyl residues
Anti-inflammatory[90]
Yam polysaccharides4.2 × 104 DaGlc:Man:Gal = 1:0.37:0.11 (1→3)-α-Glucopyranose as the main chain and β-galactopyranose-[(1→2)-α-mannopyranose]3-(1→2)-α-mannopyranose-(1→6) as the side chain Immunomodulatory[91]

3. General Rules of HPS-DDS

3.1. Preparation of HPS-DDS

In some cases, HPS can be transformed into nanoparticles through nanoprecipitation or nanosuspension [92]. However, HPS often need to be combined with other polymeric materials and drugs to prepare DDS. The strategies for combining HPS with polymers/drugs mainly include physical modification [93], chemical modification [94], electrostatic adsorption [56], and covalent linking [95] (Table 2).

3.1.1. Self-Aggregation

When HPS serve as therapeutic agents, they can directly be transformed into NPs via self-aggregation without using additional copolymers [96]. Microfluidic technology appears to be a promising strategy for the preparation of HPS NPs due to its well-controlled, reproducible, and high-throughput nature [97,98]. Microfluidic systems consist of two infusion syringe pumps and a micro T-junction, which are linked by a capillary. To prepare NPs, HPS are dissolved in an aqueous solution to obtain a separate phase, while an anti-solvent, such as acetone, serves as the continuous phase. The two phases come together at the Micro-T junction owing to the action of the infusion pump. Accordingly, the polysaccharides experience a change in solvent at the micro scale. The polysaccharide unimers transform into nuclei under the high shear force between the two phases. These nuclei grow in size through a diffusion-limited process, and resulting in the formation of “kinetically locked” NPs. By adjusting the flow rate and ratios of the solvent and antisolvent, the particle size of the NPs can be controlled [99].

3.1.2. Polymer Encapsulation

In general, polymeric NPs, such as poly(lactic-co-glycolic acid) (PLGA) and polylactic acid NPs, are synthesized using precipitation/solvent exchange/emulsification methods [100,101]. The double emulsion technique is commonly adopted to prepare NPs with polysaccharides and biodegradable polymeric materials [102].
For NP preparation, HPS are dissolved in an aqueous solution while the polymers are dissolved in dichloromethane; these solutions serve as the internal water phase and organic phase, respectively. The primary emulsion (water-in-oil) is obtained by completely mixing the two phases. Then, the primary emulsion is poured into the external water phase, and an emulsifier is added to generate a stable w/o/w emulsion [103]. After the removal of the organic solvent and centrifugation, NPs are obtained. Commonly used emulsifiers include F68 [104], F80 [105], Tween 20 [106], and triglycerides [107]. During the preparation process, the key factors that influence the quality of the NPs are the ratio of the organic phase to the internal water phase, the ratio of the external water phase to the organic phase, and the concentration of the emulsifier [108].
Polysaccharide NPs can also be synthesized using the microfluidic method. One approach involves the preparation of drug-loaded polysaccharide NPs that serve as the core and are coated with a shell made up of a polymer such as alginate [97]. In the other approach, a hydrophobic polymer, such as poly(D,L-lactide) (PLA), serves as the core, while an amphiphilic polysaccharide is employed as a stabilizer and surface modifier. Polysaccharide-covered NPs are prepared using a continuous emulsion/solvent diffusion method by employing a microfluidic flow-focusing junction [109].

3.1.3. Covalent Link

Amphiphilic polysaccharide derivatives are widely known to self-assemble or aggregate in an aqueous phase. Briefly, polysaccharide-based micelles consist of three fundamental elements: the hydrophilic domain of the polysaccharides; the hydrophobic domain of the polymer; and the linkage between the hydrophilic and hydrophobic segments (Figure 1).
In general, there are two strategies for the synthesis of amphiphilic polysaccharide derivatives [110,111]. One involves the grafting of hydrophobic polymers with functional groups throughout the polysaccharide chains. In contrast, the other involves grafting the polymers onto polysaccharide terminal groups, generating “grafted” or “block” (or “block-like”) polysaccharides derivatives. For “grafted” structures, the synthetic polymers can be directly introduced through a “grafting onto” or “grafting from” approach. For “block” structures, polysaccharide derivatives are typically obtained via the “end-to-end coupling” of a preformed polymer and the polysaccharides.
The inherent properties of amphiphilic polysaccharide derivatives, such as their Mw, degree of grafting, and volume ratio of hydrophilic/hydrophobic domains, greatly influence the physicochemical and biological properties of the derived NPs [112]. In particular, the degree of substitution (DS) of hydrophobic segments plays a key role in the quality of the micelles. As a general rule, a higher DS value is associated with a stronger hydrophobic force, which makes it easier for amphiphilic polysaccharides to self-aggregate into micelles. This results in a lower critical aggregation concentration(CAC) [113]. However, polysaccharides with too many hydrophobic modifications may precipitate in an aqueous solution, which reduces their drug loading capacity and encapsulation efficiency (EE) [114].
In addition, the linkage between HPS and hydrophobic segments is important for controlled drug release. This linkage is usually composed of stimuli-responsive bridges. Accordingly, these NPs disassemble in response to changes in internal and/or external stimuli, leading to the controlled release of loaded drugs [115,116].
Grateloupia livida polysaccharides-SeNPs are sometimes prepared using the selenite/Vc chemical reduction method by dissolving the polysaccharides in a redox system containing selenite and ascorbic acid [117]. The polysaccharides are capped onto NPs and produce a smoother NP surface. The particle size ranges from 50 to 200 nm, and the NPs show high stability and re-dispersion properties.

3.1.4. Crosslinking

In order to meet the special needs of medical application, HPS can be crosslinked to obtain three-dimensional hydrogels. In physically crosslinked hydrogels, different chains are linked through hydrogen bonding, ionic bonding, or an associative polymer–polymer interaction [118]. In contrast, in chemical crosslinking, covalent bonds/hydrogen bonds are generated between the polysaccharide and amino or carboxyl groups in other polymers by means of metal coordination [119] and the Schiff bases reaction [24]. Due to the formation of covalent bonds, chemical crosslinking provides greater resistance to environmental changes and stronger mechanical strength and stability than physical crosslinking.
HPS-based metallic NPs (HPS-MNPs) are prepared through a crosslinking strategy. In general, the HPS are dissolved in an aqueous phase and added to metal salts, with continuous stirring at a specified temperature. The particles are precipitated after the addition of a large volume of ethanol and kept on an ice-bath overnight. After centrifugation and alcohol volatilization, the sediment is dissolved in water and then lyophilized to obtain HPS-MNPs [56,120]. The three key factors that influence the quality of the HPS-MNPs are the ratio of the polysaccharides solution to the metallic salt solution, temperature of the reaction, and pH of the reaction mixture [121,122,123].
Crosslinking strategies can also be used to prepare polysaccharide NPs without adding polymers [124]. In some cases, sodium trimetaphosphate (STMP) is employed as the crosslinking agent [125]. In these cases, the polysaccharides need to be modified to provide reaction sites for the crosslinking agent. Araucaria heterophylla polysaccharides [126] and Prosopis chilensis L. polysaccharides [127] can be formulated into NPs using this strategy. Before preparation, these polysaccharides can be carboxymethylated and then reacted with STMP to form NPs. These polysaccharides can be fabricated into curcumin-loaded nanocarriers with a size below 200 nm. The drug-loaded nanocarriers show both antibacterial and anti-cancer effects, along with a steady drug release profile [128].

3.1.5. Physical Mixture

Physical modification is the most commonly used method to prepare HPS-DDS via non-covalent interactions between HPS and other polymers/drugs [129]. When mixed with other polymers, HPS can reduce viscosity and enhance mucosal adhesion. In addition, the carboxyl and hydroxyl groups provide hydrophilicity and degradability to the system [130]. For example, to prepare HPS microneedles, other polymers such as HA [49], CS [131], and alginate [132] are usually selected to improve the mechanical strength and skin puncture ability. In hydrogels, BPSs and carbomer are co-dissolved in an aqueous phase to form stable supramolecular aggregates and a cross-linked network structure [133]. Such a HPS–polymer mixture can significantly improve the colloidal strength and viscoelasticity of hydrogels [134].

3.1.6. Electrostatic Adsorption

Some cationic polymers, such as CS [135] and polyethyleneimine (PEI) [135], can form nano-complexes with HPS through simple electrostatic interaction. In one study, to improve their intestinal absorption efficacy, APSs were mixed with amphiphilic CS to form nanocomplexes with a positive zeta potential and size of 100–150 nm. These nanocomplexes showed high permeation through intestinal enterocytes [136]. In another study, a CS-tamarind gum polysaccharides (TGP) [137] polyelectrolyte nanocomplex was fabricated to stabilize simvastatin and enhance its anti-tumor efficacy. The formulated NPs showed high simvastatin entrapment efficiency (67–83%) and a small size (53–383 nm). To improve water absorption and accelerate blood coagulation, alginate and BSPs were mixed with a CS solution to generate a CS/Alg/BSPs nano-complex via lyophilization [138].

3.2. Role of HPS in DDS

3.2.1. Drug Carriers

HPS may serve as drug carriers in DDS, such as nanoparticles [139], HPS-drug complexes [140], hydrogels [141], and microneedles [142]. In such cases, HPS are introduced as a bio-copolymer or matrix to interact with the drug via absorption, conjugation, encapsulation, etc. [143]. Thus, biodegradability [10] and the compactivity between the HPS and drug [144] are the two key properties controlling drug release. The molecular weight [145], chain length [146], branches [147], charges [148], and special functional groups [149] of HPS affect the performance of the DDS. HPS with a low Mw can reduce the particle size of the NPs as well as the drug encapsulation efficiency, but increase the drug release rate [150]. In HPS-hydrogels, the chain length and branch properties significantly influence gel characteristics such as adhesion, mechanical strength, and rheological features, producing different drug release profiles [151].

3.2.2. Macromolecular Drugs

As mentioned above, HPS produce immunomodulatory effect. When introduced into DDS, HPS can serve as both a drug carrier and a macromolecular drug for immunomodulation [32]. Hence, the simultaneous and synchronous delivery of bio-macromolecules and small drugs can be achieved through HPS-DDS. In this way, HPS-DDS provide more synergistic therapeutic effects through multiple levels and mechanisms [152]. Furthermore, in some HPS-based nano formulations, the in vivo stability and bioavailability of HPS can strongly be enhanced through molecular modification [153]. In previous studies, NPs were prepared using American ginseng polysaccharides for enhanced immunotherapy effects. Lepidium meyenii Walp. (maca) polysaccharides (MP) [154] and BSPs [26] were fabricated into micelles to prepare a co-delivery platform for the polysaccharides as well as small therapeutic agents. In this way, the specific targeting and cell absorption of the bio-macromolecules could be achieved. In another study, liposomes based on Glycyrrhizophores polysaccharides (GPSL) [155] and Ophiopogon japonicus polysaccharides [156] were found to show better immunomodulatory activities than the two HPS alone. In addition, the stability of Ganoderma lucidum polysaccharides could be improved significantly after their introduction into microcapsules [157].

3.2.3. Adjuvants

With regard to immunotherapy, HPS are excellent immune adjuvants. They can: improve the levels of serum antibodies and promote the production of inflammatory factors such as IL-2, IL-4 and IL-6; promote the proliferation of lymphocytes; improve the abundance of CD4+ and CD8+ T cells in the peripheral blood; and thereby produce enhanced humoral and cellular immune responses [96,158]. Therefore, in HPS-based DDS, the polysaccharides often act as adjuvants and amplify the effects of vaccines [159]. For example, ASPs [160] and Chinese yam polysaccharides (CYP) [161] are used as adjuvants and co-encapsulated into PLGA NPs with ovalbumin (OVA). Owing to the presence of HPS, the immune responses induced by OVA are enhanced, as indicated by the higher ratio of CD4+ and CD8+ T cells as well as the long-term IgG immune response with a mixed Th1 and Th2 response.

3.2.4. Targeting Agents

Many HPS are rich in galactose and mannose structural units and can specifically recognize the highly expressed asialoglycoprotein receptor (ASGPR) [162] and CD44 receptor [163] in tumor cells, as well as MRs [164], CD206 [165], and TLR-2 [166] in tumor-related macrophages. Hence, when incorporated into DDS, the HPS may serve as targeting agents for tumor cells and TME. For example, hepatoma cells can be specifically recognized by galactose, N-acetylgalactosamine, and glucose owing to the overexpression of surface ASGPRs [167]. Thus, given the large amount of galactose and glucose present in their structures, ASPs [168] and BSPs [169] can target liver tissue. Previously, ASPs and BSPs-based DDS have been developed for the targeted delivery of drugs to hepatoma cells. MP [170] have high levels of mannose, which shows specific affinity towards tumor-associated macrophages (TAM). Thus, they were used to deliver drugs to TME in our previous study. Gracilaria lemaneiformis polysaccharides (GLP) [171], which have a high binding affinity for the αvβ3 integrin overexpressed in glioma cells, have been used to prepare GLP–SeNPs DDS for glioma targeting.

3.2.5. Stabilizers

In recent years, numerous studies have reported that HPS can enhance the stability of nanosystems across a wide range of pH values and temperatures, and improve cellular uptake owing to their hydrophilic hydroxyl groups and high specific surface area [172]. In one study, the solubility of quercetin was increased by 68.88-fold after its covalent linkage to APSs, leading to enhanced stability. This was because the association constant between quercetin and the polysaccharides was high, resulting in a large solubilization coefficient in the system [173]. In SeNP systems, citrus lemon polysaccharides were used as modifiers and stabilizers [174]. With its high stability and good dispersion in water, the nanosystem showed significant antitumor effects against HepG2 cells. In HPS-Metal NPs (HPS-MNPs) [175] and HPS-inorganic NP systems [176], HPS possess excellent biocompatibility and provide effective transportation for MNPs in vivo. The presence of abundant -COOH and -OH groups in HPS can improve the solubility and stability of MNPs, resulting in a longer circulation time [177]. The polysaccharides reduce the toxicity of MNPs and increase their therapeutical effects and degradability. In addition, Polygonatum sibiricum polysaccharides [178] and APSs [179] have been introduced into SeNPs as stabilizers and dispersing agents to reduce their surface energy through hydrogen bonding, hydrophobic interaction, and electrostatic interaction.

3.2.6. Emulgators

As a form of hydrophilic colloids, HPS have good emulsifying and thickening effects. Therefore, they are considered excellent emulsion stabilizers and are widely used in the preparation of emulsions [180]. In general, for the preparation of polysaccharide-emulsions, Tween-60, Tween-80, Span-60, and Span-80 are used as surfactants; soap, sodium lauryl sulfate, polyol fatty acid esters (such as monostearate glycerides), and polysorbates are mainly used as emulsifiers; and polyethylene glycol ethers, liquid paraffin, glycerin, soybean oil, olive oil, and stearyl alcohol are commonly used as the oil phase [181]. In particular, flavonoid-grafted soybean polysaccharides can emulsify unstable oil-in-water emulsions [182], and soy CS can be used to develop emulsion food delivery systems [183]. Albizia lebbeck L. polysaccharides (ALPS) are hydrophilic non-ionic polysaccharides. Owing to unique functional properties such as high thermal stability and pH sensitivity, ALPS show good potential as natural emulsifiers [181].

3.2.7. Solubilizers

The solubilization effect of HPS is attributed to their specific structure and hydrophilic groups. The weakened intramolecular and intermolecular hydrogen bonds in HPS chains lead to low crystallinity and better water solubility [184]. Studies have shown that vinegar baked Radix Bupleurum polysaccharides (VBCP) can increase the solubility of baicalin and rhein. As natural surfactants, VBCP can self-assemble in water to form micellar aggregates, which can encapsulate insoluble drugs through the interaction of hydrogen bonds and hydrophobic forces [34].
Table 2. Summary of HPS-DDS.
Table 2. Summary of HPS-DDS.
Type of Dosage FormPolysaccharidesDrugCuring DiseasePreparation MethodThe Role of HPS in DDSAimed CellsSize (nm)Zeta (mV)Refs
HPS nanoparticlesPanax quinquefolium polysaccharides Immunoregulation; Reduce skin cancerSelf-aggregation; Polymer encapsulationImmunologic adjuvant; Macromolecular drugsMacrophages180 ± 10; 20 [99,185]
HPS–polymeric nanoparticlesRamulus mori polysaccharides Inflammatory bowel disease; ColitisPolymer encapsulationMacromolecular drugsMacrophages205.6 ± 4.26;
180.3 ± 4.21
−31.7 ± 1.097[102,104]
Inulin AntigensStimulated the Th2 type immune response.Polymer encapsulationImmunologic adjuvantAntigen presenting cells
(APC)
1.5 ± 0.12 [105]
Aloe polysaccharides Angular leaf spotPolymer encapsulationMacromolecular drugs 644.00 ± 0.52; 243.20 ± 0.22 [106]
Chinese yam polysaccharidesOvalbuminStrengthen immune responsesPolymer encapsulation; Covalent linkImmunologic adjuvantCD3(+)CD4(+) T cells CD3(+)CD8(+) T cells200  [186]
Angelica sinensis polysaccharidesOvalbumin; Inactivated H9N2Induce strong and long-term immune responses; H9N2 influenzaPolymer encapsulationMacromolecular drugs; Immunologic adjuvantCD4(+)/CD8(+) T cells
Th1 cells
225.2;
286.3 ± 2.45
−11.27; 47.8 ± 0.24[187,188]
Dendrobium polysaccharidesOvalbuminImprove immune responsesPolymer encapsulationImmunologic adjuvantMacrophages and lymphocytes
CD4(+)/CD8(+) Tcells
141.4; 156.4; 175.9 −17.9 ± 1.29; −26.9 ± 2.76; 31.4 ± 2.18[189]
MicellesLepidium meyenii Walp(maca) polysaccharidesChloroquineCancer immunotherapySelf-aggregationTargeting; Immunologic adjuvant4T1-M2 macrophages120−35[154]
Angelica sinensis polysaccharidesCurcumin;
Doxorubicin
Acute alcoholic liver damage; Liver cancerPolymer encapsulation; Covalent linkDrug carrier; TargetingHepG2208.4; 228−20; −17[162,168]
Bletilla striata polysaccharidesDoxorubicin; Docetaxel; Silymarin; Let-7b; AlendronateAntitumor; Liver diseases; Suppressive tumor microenvironment; Suppressed tumor progressionCrosslinking; Covalent linkDrug carrier; Targeting; StabilizerHepG2, HeLa, SW480, and MCF-7
HepG2 cell lines
Michigan Cancer Foundation-7 (MCF-7) cells
Dendritic cells (DCs)
Macrophages
125.30 ± 1.89; 96.27 ± 1.21; 96.54 ± 5.27; 99.21 ± 3.83; 121.61 ± 9.81; 125.30 ± 1.89; 120; 67−26.92 ± 0.18; −35.66 ± 0.28; −35.46 ± 0.10; −34.76 ± 0.22; −28.37 ± 0.12; −26.92 ± 0.18; −13; −19[169,190,191,192,193,194,195]
Rehmannia glutinosa polysaccharidesInactivated BbBordetella bronchiseptica (Bb)Crosslinking; Covalent link; Self-aggregationImmunologic adjuvantDendritic cells (DCs)
CD4(+) and CD8(+) T-cells
[196]
Astragalus membranaceus
polysaccharides
Inhibited the growth of tumorSelf-aggregationImmunologic adjuvantDendritic cells (DC)
CD4(+) T/T-reg and CD8(+) T/T (reg)
138 ± 5−12.4 ± 0.3[197]
Polysaccharide-drug conjugationsLycium barbarum polysaccharidesPlatinum-based; DoxorubicinAnticancerCovalent linkTargeting; Immunologic adjuvant; Stabilizer; Immunologic adjuvantA549 (human lung cancer cell line)
human Hepatic cancer cell line HepG2
273.3 −25.6 [21,198]
Polygonum multiflorum polysaccharides5-fluorouracilAntitumorCovalent linkTargeting; Immunologic adjuvantSplenocytes and peritoneal macrophages124.7 [199]
Polysaccharide-MetalRosa roxburghii polysaccharidesAgNPsAntibacterialCrosslinkingStabilizer 24.5-83.2−36[200]
Astragalus membranaceus polysaccharidesAgNPs;
AuNPs
Antibacterial; Antitumor and immunoregulationCrosslinkingStabilizer; Stabilizer; Immunologic adjuvantDendritic cells/T cells65.08; 25.38−28.33[120,201]
Dioscorea opposita Thunb polysaccharidesZnNPsAnti-diabetesCrosslinking; Polymer encapsulationStabilizer; Immunologic adjuvant [202]
Leucaena leucocephala Seeds polysaccharidesAgNPsAnticancer, Antifungal and PreservativeCrosslinkingStabilizer; Solubilizer 8–20−14.2[203]
Tamarind polysaccharidesAgNPs;
AuNPs
Antibacterial; Anticancer and immunomodulatoryCrosslinking; Polymer encapsulationStabilizer; Immunologic adjuvant 44–86; 30–40; 20−36.7[204,205,206]
Farfarae Flos polysaccharidesAgNPsAnticancerSelf-aggregationStabilizerHT29 cells4–25−17.1[207]
Psidium guajava L. leaf polysaccharidesAgNPsAntioxidation or antimicrobialSelf-aggregationStabilizer 25–35−25.23[208]
Soybean polysaccharidesAgNPsAntibacterialSelf-aggregationStabilizer [209]
Moringa oleifera seed polysaccharidesAgNPsWound healingSelf-aggregationStabilizer 17.6−25.6[210]
Apple polysaccharidesAuNPsAnti-diabetesPolymer encapsulationStabilizer 124 ± 8.55−10.5 ± 0.54[211]
HPS-inorganic nanoparticlesGracilaria lemaneiformis polysaccharidesSeNPsAnticancerCovalent link; CrosslinkingSolubilizer; TargetingGlioma cells123−24.0[171]
Citrus limon polysaccharideSeNPsAntitumorCovalent linkStabilizer 85.35; 79.67; 90.14−9.44; −7.52; −6.87[174]
Polygonatum sibiricum polysaccharides SeNPsAntioxidationCovalent linkStabilizer 105−34.9[178]
Astragalus polysaccharidesSeNPs; ChitosanAntioxidation, enhance the proliferation of T-lymphocytes and Inhibit HepG2 cells proliferation; Exhibited high permeation through intestinal enterocytesCovalent link; Self-aggregation; Electrostatic adsorptionSolubilizerHepG2 cells478.1; 100–150−20.39; +16[136,179]
Lignosus rhinocerotis polysaccharidesSeNPsAntioxidationCovalent linkImmunologic adjuvant; stabilizer 50 [212]
Lycium barbarum polysaccharidesSeNPsAnti-fatigue; Antitumor;
Protect human lens epithelial cells (HLECs) from UVB-induced damage; Antioxidation
Covalent linkStabilizer; SolubilizerLens epithelial cells105.4; 111.5–117; 150–200; 83–160−37; −24.1[111,213,214,215,216]
Codonopsis pilosula polysaccharidesSeNPsInhibit the proliferation and promote apoptosis of HepG2 cellsCovalent linkSolubilizer; StabilizerHepG2 cells75 [217]
Dandelion polysaccharidesSeNPsAnti-tumorCovalent link; Polymer encapsulationImmunologic adjuvant; stabilizer;
Solubilizer
HepG2, A549, and HeLa50 [218]
Other HPS based NPsAraucaria heterophylla polysaccharidesCurcuminAnticancer; Antioxidation and antibacterialPhysical mixture; CrosslinkingSolubilizer; Targeting; Drug carrierMCF7 human breast cancer cell line250–300; 200 [126,127]
Tamarind Gum polysaccharidesSimvastatinAntitumorCovalent link; Electrostatic adsorptionSolubilizer; Targeting; StabilizerHuman breast cancer cell line53.3–383.1 [137]
HydrogelBletilla striata polysaccharidesLactobacillus plantarum probioticsSkin lesions; Bleeding disorders; Wound infectionCovalent link; CrosslinkingImmunologic adjuvant; Stabilizer;L929 cells [30,219,220]
Plantago psyllium seed polysaccharides5-fluorouracilAntitumorCrosslinkingImmunologic adjuvant; stabilizer; [221]
Tamarindus indica polysaccharidesSilver nanoparticleWound infectionCrosslinkingStabilizer [222]
MicroneedlePanax notoginseng polysaccharidesDoxorubicin and 5-fluorouracilAntitumorPhysical mixtureDrug carriers; Immunologic adjuvantSkin dendritic cell [49]
Bletilla striata polysaccharidesOvalbumin; Triamcinolone acetonide and verapamilInfectious disease; Hypertrophic scarsPhysical mixtureStabilizer; Drug carriers; Immunologic adjuvant [223,224]
HPS-based liposomeCordyceps sinensis Sacc polysaccharides Polymer encapsulation [225]
Lycium barbarum polysaccharides Immunological and adjuvanticity against PCV2 in vivoPolymer encapsulationImmunologic adjuvant; Macromolecular drugsSpleen cells, macrophagesCD4(+)/CD8(+) T cells120.7 ± 0.84 [226]
HPS-EmulsionDioscorea opposita Thunb polysaccharides Polymer encapsulationEmulsifier 1500−30 [107]
Albizia lebbeck L. seed polysaccharides Polymer encapsulationEmulsifier 1160–2790 −35.83−−19.00[181]
Soybean polysaccharidesSoy proteinEmulsion digestion in the gastrointestinal tractPolymer encapsulationStabilizer 835−129.76 [183]

4. HPS-DDS and Their Immunomodulatory Effects

4.1. HPS-Nanomedicine

4.1.1. HPS NPs

AGPSs—which are well-known macromolecular immunomodulators—are also used to prepare polymeric NPs for enhanced immunotherapy [185,227,228]. AGPS NPs prepared using a microfluidic approach show an average size of 20 nm. These NPs are encapsulated within porous nanospheres (~180 nm) made up of biodegradable gelatin [229]. They show a concentration-dependent enhancement of immune stimulation in both cellular and animal models [227]. In addition, these AGPS NPs are also used for dermal application, as they can inhibit the ultraviolet (UV) radiation-induced imbalance in the endogenous antioxidation system owing to their anti-oxidative and anti-inflammatory properties [230,231].

4.1.2. HPS–Polymeric Nanoparticles

In previous studies, HPS with strong immunoregulatory activities, such as Ramulus mori polysaccharides (RMPs) [102,104], Amomum longiligulare polysaccharides (ALPs) [100], and CYPs [101,186], have been encapsulated into PLGA to form HPS-PLGA NPs using the double emulsion method. The obtained NPs have a spherical morphology and a narrow size distribution. In addition, the particle size varies from 100 nm to 400 nm depending on the type of polysaccharides and the preparation process used.
In polymeric NPs, the polysaccharides can serve as immunoregulatory biomacromolecules. Some HPS can downregulate the expression of inflammatory cytokines while promoting the production of IL-10 and boost the phenotypic switch from M1 to M2 in macrophages. For example, RMPs were packaged in PLGA NPs to develop a novel anti-inflammatory nanomedicine and treat acute inflammatory bowel disease by regulating macrophages and T cells [102,104]. In contrast, some HPS, such as ALPs and CYPs [186], can re-polarize M2 macrophages to the M1 phenotype and produce a strong immune-activation effect. Further, both these contrasting immune regulation activities can be enhanced through nanosizing and polymer encapsulation.
Despite their therapeutic potential, the low bioavailability and brief biological half-life of HPS have limited their clinical applications. HPS–polymeric NPs represent an effective tool for improving the in vivo stability and bioavailability of HPS. Accordingly, the immunoregulatory activities of HPS are significantly enhanced after encapsulation into NPs [100,101].
In addition, these NPs serve as a platform for the co-delivery of polysaccharides and vaccines. In these applications, the polysaccharides act as adjuvants and amplify the effect of vaccines. For example, in one study, ASPs and OVA were co-encapsulated into PLGA NPs [187]. Because of the presence of ASPs, the immune responses induced by OVA were enhanced, as indicated by the higher ratio of CD4+ to CD8+ T cells as well as the long-term IgG immune response with a mixed Th1 and Th2 response. In another study, PEI-modified CYP-encapsulated PLGA NPs (CYP-PEI) were developed as a potential vaccine delivery system to trigger strong and persistent immune responses [232].
Stimulus-responsive HPS–polymeric NPs can also be developed. ASPs-ammonium bicarbonate co-encapsulating PLGA NPs [103] were prepared in one study. Here, the ammonium bicarbonate conferred a pH-responsive effect to the delivery system. Accordingly, ASPs release was boosted.
HPS-PLGA NPs can be further developed to achieve a complex nanostructure. In earlier works, CYP-PLGA NPs and ASPs-PLGA NPs [188] were surface modified using PEI. These positively charged PEI-coated NPs enabled the delivery of the antigen and CYP immunomodulator to lymph nodes and activated DCs, further enhancing the mixed Th1/Th2 response and Th1-biased immune response in vivo. In another study [189], dendrobium polysaccharides (DP)-PLGA-OVA NPs were developed and coated with a PEI-modified macrophage cell membrane. The macrophage cell membrane enhanced the immune effectiveness, while the PEI provided a positive charge and improved cell adsorption. Accordingly, biomimetic nano-vaccines can promote antigen uptake by macrophages and enhance lymphocyte proliferation.

4.2. HPS Amphiphilic Derivatives

4.2.1. HPS-Based Micelles

The development and applications of polysaccharide micelles as drug carriers are widely reported, BSPs are a good example. Certain hydrophobic polymers can be grafted onto the BSPs to form amphiphilic polysaccharides-copolymer derivatives using compounds such as cholesteryl [233], stearic acid (SA) [113,169,190,191], fatty acids [110].
The prepared amphiphilic polysaccharide derivatives can be self-assembled into micelle structures with particle sizes of 100–300 nm depending on the proportion of the hydrophilic and hydrophobic domains. Given their good performance in hemolysis and cytotoxicity analyses, these micelles can be used as drug carriers. Hydrophobic drugs, including docetaxel [191] and doxorubicin (Dox) [26,113,192], can be encapsulated into the hydrophobic core of these micelles. In addition, other HPS, such as APSs [162,168], and Rehmannia glutinosa polysaccharides (RGPSs) [196] can also be used to prepare micelles with the “grafted onto” strategy.
Through further structural modification, stimuli-responsive properties can be added to the micelles. Typically, two strategies are used for such modifications. In the first strategy, functional groups are directly introduced onto the polysaccharide chain. In our previous study, histidine (His)- and SA-modified BSPs derivatives were synthesized. The prepared micelles showed stepwise pH sensitivity due to the protonation-deprotonation effects of the imidazole group in His. Therefore, the prepared BSPs micelles showed boosted drug release under acidic conditions [26,192]. In the second strategy, the functional group is used as a linker. For example, one study used cystamine (CYS) to link BSPs and SA. As the CYS linker could be broken down under reducing conditions, the micelles could be disassembled, resulting in a fast drug release profile [234]
Micelles can also be modified to achieve targeting effects. In a previous study, a folate-and SA-modified BSPs (FA-BSPs-SA) polymer was synthesized. The micelles could enter tumor cells through folate receptor-mediated endocytosis via a clathrin-dependent mechanism. This facilitated the synergistic anti-tumor effects of the loaded Dox in 4T1 cells, resulting in enhanced anti-tumor effects [193].
Owing to their metabolic features, HPS micelles possess some unique properties. It has been reported that hepatocellular carcinoma cells can specifically recognize various ligands such as galactose, N-acetylgalactosamine, and glucose via the ASGPR expressed on their surfaces [235] Thus, ASGPR-mediated DDS have gained interest in the field of hepatocellular carcinoma treatment. It was demonstrated that some HPS, such as BPSs and ASPs, show a high affinity to ASGPR due to their higher galactose content, branched structure, and appropriate spatial geometry. Hence, BSPs and ASPs micelles possess liver-targeting capabilities and are promising nano carriers for drug delivery to the liver [168,169]. In addition, the specific glycosidic linkages of polysaccharides can also confer targeting properties. BSPs contain β-glucose and α-mannose at a relative molar ratio of ~2.4:1. MRs are strong lectins that can recognize and bind to BSPs. Due to the overlapping expression of MRs on TAMs and tumor-infiltrating dendritic cells (TIDCs), BSPs nano vehicles can be used as targeted DDS for tumor immunotherapy [194,195].
HPS micelles can also be used as a co-delivery platform for bioactive macromolecules and small therapeutic agents. In a previous study, we fabricated TAMs-targeting amphiphilic polysaccharide micelles for synergistic cancer immunotherapy. We grafted a PLGA segment onto MP, which are naturally derived macromolecules with a strong TAMs-remodeling effect. Disulfide bonds were used as a redox-sensitive linkage. The amphiphilic polysaccharide derivatives could self-assemble into core (PLGA)-shell (MP)-structured micelles and encapsulate chloroquine (CQ) into the hydrophobic core. After administration, the micelles co-delivered MP and CQ to TAMs and achieved a synergistic immunotherapeutic effect in tumors via the multiple regulatory effects of MP and CQ. Accordingly, the targeted delivery of active macromolecules was achieved [154]. In another study [197], the hydrophobic compound 4-(n-octyloxy) phenylboronic acid was simply conjugated to APSs. The amphiphilic polysaccharide derivatives could self-assemble into NPs (ANPs). These ANPs could reverse TEM and thereby enhance the radiation-induced abscopal effect. A mechanistic study demonstrated that the ANP-induced immune response was mainly mediated by DCs activation, manifesting as phenotypic maturation and enhanced antigen presentation through the TLR4 signaling pathway.

4.2.2. HPS-Drug Conjugations

Polymer-drug conjugates (PDC) have exhibited clinical and commercial success in both drug delivery and targeted treatment for cancer, diabetes, arthritis, and pain [236,237,238]. At present, polysaccharides-drug conjugates (PSDCs) appear to be effective options and have attracted considerable attention. PSDCs are simply prepared by covalently linking a drug to the functional groups in the polysaccharides. During the preparation process, the reaction time, reaction temperature, amount of catalyst, and molar ratio of polysaccharide to drug affect the drug loading activity [239].
HPS can also act as bioactive agents. Some immunomodulators—such as ASPs, Lycium barbarum polysaccharides (LSPs), Polygonum multiflorum polysaccharides (PMPSs), and APSs—were employed to construct PSDCs [34]. The PSDCs enabled the co-delivery of these bioactive macromolecules with therapeutic agents such as dexamethasone (Dex), platinum-based antineoplastic drugs, Dox, 5-fluorouracil, and ibuprofen, among others [240,241]. Due to their self-assembly characteristics, the prepared PSDCs possessed the specific features of NPs, showing selective accumulation in reticuloendothelial system phagocytic cells as well as tumor cells. The LBPs-Dox complex showed the strongest inhibition against the proliferation of tumor cells, higher than that of polysaccharides and the drug alone, indicating that LBPs and Dox exerted a synergistic anti-tumor effect [21]. When 5-fluorouracil was covalently linked to PMPSs, the levels of IL-2 and TNF-α were further upregulated. Thereby, the immunosuppression in TME was re-sharped, and the anti-tumor effects were enhanced [199]. The ASPs-Dex conjugate has also been shown to exert a synergistic effect in the treatment of ulcerative colitis in rats. The ASPs limit the overall amount of drug absorbed and thus reduce the systemic immune suppression caused by Dex alone [242].
In addition, the high fractions of mannose and glucose in PSDCs enable targeted delivery to the liver by resident macrophages and sinusoidal endothelial cells. Due to the overlapping expression of MRs on TAMs and TIDCs, cationic BSPs (cBSPs), which contains high amounts of mannose moieties, can be used to conjugate therapeutic agents such as bisphosphonate and let-7b and efficiently target and specifically re-shape TME for cancer immunotherapy [194,195]

4.3. Polysaccharides-Metal Nanoparticles

Metallic NPs (MNPs) are promising materials for DDS [243]. HPS have been recognized as valuable agents for the synthesis of MNPs owing to their outstanding biocompatibility, biodegradability, and targeting properties [244].
Generally, HPS-MNPs exhibit a “core-shell” structure [202]. The core is composed of metallic NPs (Pt, Au, Ag, Zn, or their oxides), while the polysaccharides are coated onto their surface, acting as both stabilizing and reducing agents and adhering to the MNPs through noncovalent bonding [203,204,207]. The carboxyl and hydroxyl groups of the polysaccharides play a key role in mediating the solubility and stability of the MNPs in the HPS-MNP complex, preventing the flocculation and aggregation of MNPs [245]. As a result, the HPS-MNPs complex can form a stable suspension that carries a negative electrostatic surface charge (Figure 2). Moreover, the particle size ranges from 10 to 150 nm [201,246].
In some cases, HPS such as APSs [120] and carboxymethyl tamarind polysaccharides (CMTs) show synergistic antibacterial effects along with MNPs. It has been reported that CMT AgNPs (CMT-AgNPs) exert anti-bacterial effect without damaging immune cells such as macrophage and keratinocytes. Hence, they hold outstanding promise in the treatment of bacterial infections in vivo [205]. Other polysaccharides, such as Rosa roxburghii Tratt fruit polysaccharides (RP3) [200], and soluble soybean polysaccharides (SPSSs) [209], do not have significant antibacterial effects when used alone, but can strongly enhance the activities of MNPs, enabling sustained release with long-term use.
During the healing process, the polysaccharides can regulate macrophages, which play an important role in cell proliferation, vascularization, collagen deposition, and granulation, by secreting various chemokines and cytokines. Moringa oleifera seed polysaccharides-embedded silver NPs (MOS-PS-AgNPs) show a greater bactericidal effect and lower toxicity than plain AgNPs. In addition, the complex promotes wound healing by upregulating IL-10 and downregulating IL-6 expression [210].
Further, PMNPs can target different tumor cells and treat various types of cancers depending on the polysaccharides and MNPs used. The polysaccharides PST001 are isolated from the seed kernels of Tamarindus indica (PST), and PST-Gold NPs exert anticancer effects through the induction of apoptosis and increase the count of CD3/4/8 cells in the bone marrow [206]. One study showed that ASPs-AuNPs can increase not only NO release in the culture environment of DCs but also enhance the gene expression of DCs-derived cytokines, promoting the proliferation of CD4+ and CD8+ T cells in splenocytes [201]. The Farfarae Flos polysaccharides-Ag complex (FFP@AgNPs) significantly decreases the proliferation ability of HT29 tumor cells, inhibits their migration, and promotes their apoptosis [207]. In LBPs platinum-based conjugates (LBPs-5-ASA-Pt), Pt shows better binding to DNA and forms Pt-DNA adducts, leading to greater nuclear accumulation and irreversible lesions in the double helix, which ultimately result in tumor cell death [247].
Further, HPS-MNPs have been developed as new potential candidates for the treatment of diabetes mellitus (DM) and its complications. Modified apple polysaccharides (MAP)-AuNPs were conjugated with insulin (INS) for the oral treatment of type 1 DM [211] while a Dioscorea opposita Thunb. polysaccharides-zinc (DPS-Zn) inclusion complex was synthesized for the treatment of type 2 DM [202]. In this formulation, the HPS can restore the immune balance by regulating cytokines and enhancing the body’s immunity, producing synergistic treatment effects against DM.

4.4. HPS-Inorganic Nanoparticles

Selenium (Se) is an essential micronutrient that exerts its biological effects mainly via selenoproteins. SeNPs have functional characteristics such as a high absorption rate, low toxicity, and high biological activity. Therefore, they are widely used in health foods and the field of medicine. However, SeNPs have the disadvantages of a high surface energy and poor stability. They easily form grey and black elemental selenium and lack selectivity against cancer cells. Considering that both HPS and SeNPs possess unique physicochemical characteristics and bioactivities, it is reasonable to assume that HPS-SeNPs would have their combined advantages and show stronger biological effects, such as anti-tumor, anti-oxidation, and immunomodulatory effects. Thus, they could potentially be used as nutritional Se supplements in food and medical applications.
When LBPs are used as the capping agent, LBPs-SeNPs exhibit better stability [141], with enhanced bioactivities, including anti-fatigue [214], anti-tumor [111], UV Protection [215], and antioxidation effects [216]. Rosa roxburghii fruit polysaccharides (RTFP)-SeNPs exert synergistic protective effects against H2O2-induced apoptosis in INS-1 cells. In this formulation, RTFP not only serves as the stabilizer but also exhibits high antioxidation and α-glucosidase-inhibiting effects. Inulin, a fructan-type HPS widely distributed in nature, has been demonstrated to have prebiotic effects and anti-tumor, anti-oxidation, and immunomodulatory activity. In a previous report, inulin fructans from Codonopsis pilosula [248], T. mongolicum [218] as well as Citrus limon (L.) Burm. f. (Rutaceae) [174] were introduced into SeNPs for tumor treatment. In addition, Ulva lactuca polysaccharides-SeNPs were found to potentially attenuate colitis by inhibiting NF-κB-mediated hyperinflammation.

4.5. HPS-Based Hydrogels

Hydrogels are physically or chemically cross-linked hydrophilic polymers that can immobilize large amounts of water or biologic fluids into their 3D structures [249]. The polymeric network in hydrogels consists of hydrophilic polymer chains that are linked to each other via physical and chemical crosslinking [250]. HPS can be introduced into hydrogel systems to improve their biocompatibility. Polysaccharides in hydrogels can play three major roles. First, they can act as a pharmaceutical agent and exert independent therapeutic effects. Second, they can enable more functional modifications and respond to a greater variety of biological environments. Finally, the reducing groups of polysaccharides can improve the stability of metal NP-hydrogel composite systems and play a protective role.
The simplest approach for hydrogel preparation involves physical cross-linking (one-pot method). Currently, BSPs and APSs are commonly used to prepare hydrogels with good biocompatibility and mechanical strength via physical crosslinking. A Snakegourd root/Astragalus composite OPCH hydrogel was successfully printed into three differently shaped patches using a melt extrusion 3D printer [89]. Previously, we prepared a BSPs hydrogel using the one-pot method with carbomer as the matrix. In another work, BSPs was mixed with the hydrophilic monomer acrylic acid (AA) [30], and the linear polymer polyvinyl alcohol (PVA) was introduced to create a double network [219]. To enhance the mechanical properties of the hydrogel system, CS was reacted with oxidized BSPs (OBSPs) [220], resulting in greater intermolecular bonding and better wound healing activity [251] from an immune regulation perspective.
Furthermore, chemical- and radical-induced free radical polymerization can also be used to prepare HPS-hydrogels. For example, the graft copolymer hydrogels and three-dimensional interpenetrating networks (IPNs) of Psyllium polysaccharides (Psy) and methacrylic acid (MAAc) were prepared using ammonium persulphate as the initiator and N,N-methylenebisacrylamide (N,N-MBAAm) as the crosslinker [252,253]. In addition, the irradiation of polymers in a suitable system is also useful for preparing hydrogels without any chemical initiator/cross linker [254]. Psyllium-N-vinylpyrrolidone (NVP)-based hydrogels (psy-cl-poly [NVP]) were prepared through radiation-induced crosslinking [221]. The polymerization involved the irradiation of an aqueous Psyllium polysaccharides solution, resulting in the generation of a significant number of radicals on polymer chains [221].
In particular, composite polysaccharide hydrogels loaded with metal NPs have also received attention. In these systems, the metal NPs act as antibacterial agents, while the HPS serve as a carrier for the metal NPs and reduce toxicity and promote wound healing. Galactoxyloglucan (PST), a HPS isolated from the seed kernel of Tamarindus indica, was used to prepare hydrogels with Au/Ag NPs [222].

4.6. HPS-Based DMNs

Dissolving microneedles (DMNs) have been widely studied for their applications in transdermal drug delivery because they can dissolve within the skin [223]. HPS are extensively used for the fabrication of DMNs due to their biocompatibility, biodegradable nature, and sustainable delivery. The most widely explored polysaccharides for DMN-related applications include HA, dextran, CS, cellulose, sodium alginate (SA), and blends of other biopolymers [255]. Compared with ordinary polymers, HPS not only offer a wider range of functional groups, good biocompatibility, biodegradability, and other polymer-related properties, but also have immunomodulatory action (Figure 3).
BSPs microneedles (BMNs) are the most widely studied. Mechanically robust simple BMNs are prepared using centrifugation [142,223]. The prepared BMNs exhibit better mechanical properties and stability than microneedles made of HA and polyvinyl alcohol. BMNs can dissolve in the interstitial fluid of the skin after insertion and release the encapsulated OVA to prime the immune system. These encouraging findings indicate that BMNs can be a promising tool for effective vaccine delivery. Further, the micro-molding method can be used to prepare more complex forms of MNs with carboxymethyl CS (CMCH) and BSPs for hypertrophic scar (HS) treatment [224]. Hydroxypropyl β-cyclodextrin (HP-β-CD) is used to encapsulate hydrophobic triamcinolone acetonide (TA), and the obtained inclusion is co-loaded with hydrophilic verapamil (VRP) in MNs. The MN-HP-β-CD(TA)-VRP MNs are then attached to an EC-based layer to obtain a MN patch, which shows a higher mechanical strength and stronger internal interaction of hydrogen bonds than observed with BSPs alone. The BMNs significantly decrease the thickness of HSs as well as their hydroxyproline (HYP) and transforming growth factor-β1 (TGF-β1) expression. Accordingly, they improve collagen fiber arrangement and reduce dermis congestion and hyperplasia.
In our previous study [49], we prepared HPS MNs using Panax notoginseng polysaccharides (PNPS) as the matrix. The PNPS MNs possess suitable mechanical strength and good solubility, enabling them to easily penetrate the SC layer and release the loaded drug into the deeper layers of skin. Upon entering the skin, the bioactive PNPS recognize and target skin DCs via TLR2 and TLR4, triggering DCs maturation and enhancing the local DCs-mediated T cell immune response. Thus, these PNPS MNs could serve as promising carriers for transdermal drug delivery and natural adjuvants for transcutaneous immunization.

4.7. Other HPS-Based DDS

4.7.1. Liposomes

The development of HPS-lipid formulations has become a hot spot of research. Liposomes can encapsulate polysaccharides in their aqueous cores and/or membranes, and the unique physical and chemical properties of liposomes not only improve the bioavailability, therapeutic effect, and dosage of polysaccharides but also enable their targeted delivery [256,257,258].
The polysaccharides commonly used in the preparation of liposomes are white mustard polysaccharides, Yu ping feng polysaccharides, wheat dong polysaccharides, APSs, and goji berry polysaccharides. In addition, valuable herb plants such as Ganoderma lucidum and Cordyceps sinensis are also used in the development of polysaccharides-liposomes. Compared with APSs, APSs-liposomes not only induce greater phagocytic activity in macrophages but also improve the ability of DCs to stimulate T cell proliferation and develop antigens [225]. Wheat dong polysaccharides-liposomes can significantly increase SOD and oxidase levels and reduce MPO levels. Further, they can significantly increase the proportion of splenocyte proliferation and CD4+ and CD8+ T cells while also enhancing cytokine secretion [259]. LBPs liposomes can act in synergy with PHA or LPS to significantly promote splenocyte proliferation, increase the ratio of CD4+ and CD8+ T cells, promote macrophage cytokine secretion; enhance the PCV2-specific IgG antibody response, and promote the secretion of IFN-γ, TNF-α, and IL-4 [226].

4.7.2. Emulsions

Some HPS, such as Albizia lebbeck L. seed polysaccharides, Dioscorea opposita Thunb polysaccharides, and APSs can be used as natural emulsifiers and smart polymers for the preparation of pH-sensitive drug delivery system emulsions [107,181]. In addition, APSs nano-emulsions can induce specific antibodies and enhance the Th1 and Th2 immune response. They can significantly enhance the body’s ability to produce antibodies and enhance the immunogenicity of antigens, improving vaccine effectiveness and reducing the immune response after vaccine administration [107].

5. Conclusions and Prospect

HPS are biodegradable, carbon neutral, environmentally friendly, and pose a low risk to human health and safety. They are easily modifiable and have other unique physical and chemical properties. Therefore, they are good carriers for DDS. HPS also have immunomodulatory effects, which further add to their advantages. Therefore, the application of HPS in DDS has become a research hotspot. In this review, a wide range of HPS-DDS were systematically summarized through formulation-based categorization. A brief introduction of some frequently studied HPS was provided, followed by strategies and methodologies used for polysaccharides-based DDS fabrication. In particular, we focused on the role of HPS in these DDS.
Many studies have been reported that HPS are rich in biological activities, safe and non-toxic side effects. However, HPS-DDS has not been deeply discussed. Although some clinical studies on HPS-DDS have been initiated, research on HPS-DDS is largely limited to the preclinical stage, and some problems need to be addressed before their clinical translation, such as the evaluation of their long-term safety, toxicological profile and the quality control standard system and so on. Further, Novel characterization and preparation techniques are required for controlling the quality of HPS-DDS and overcoming batch variations in the polysaccharides. In addition, scalable, cost-effective, and reproducible manufacturing systems need to be developed for HPS-DDS. In addition, the interactions between the polysaccharides and other components of the DDS as well as the mechanisms of polysaccharide NPs synthesis need to be investigated more thoroughly. In view of the above problems, HPS-DDS still has many potential problems before they get approval by regulators. Therefore, it is still needing the unremitting efforts of scientific researchers to establish the more perfect structure of HPS-DDS.
As a natural product, HSP were obtained by extraction and purification from herb plants. Thus, the purity was an important issue influencing the functions. For example, trace amounts of lipopolysaccharides in polysaccharides would significantly affect their immunoregulatory activities. In the relevant articles, the HSP used in DDS are obtained under complex purification process. First, the HSP should be deproteinization and decolorization in advance. Then, the HSP were purified by DEAE column to remove the general impurities (e.g., oligosaccharides, proteins and lignin). After that, they are further purified by sephadex column including G20, G50, G100 and G200 to obtain the homogeneous products with narrow Mw distribution. As a result, the products usually have an acceptable purity above 90%. In our previous work, PNPS, MP and BSPs were used to fabricate the DDS such as micelle, microneedles and hydrogel. the purities for all there HSP were all above 95% [49,154,192,260].
When designing HPS-based NPs for a specific purpose, it is crucial to control polysaccharides properties such as molecular weight, monosaccharide composition, and type of glycosidic linkage. However, unlike synthetic polymer materials, HPS are not amenable to property modifications. The region of plant production, plant age, and pretreatment methods all significantly influence the quality of HPS. In addition, preparation methods such as water extraction, alcohol precipitation, and column chromatography, also affect the properties of HPS. Therefore, the internal relationship between the preparation process and the quality of polysaccharides needs to be further explored. Moreover, integrated extraction and separation technologies need to be employed at a large scale to standardize the extraction and purification process. In this way, a large number of raw polysaccharide materials could be obtained for further investigation while controlling quality.
Numerous studies show that many HPS have significant immunomodulatory activity and can inhibit tumors by independently activating the immune response. This is a major advantage of HPS-DDS. However, the bioactivity of polysaccharides is closely related to their structure. Thus, caution must be exerted before structurally modifying HPS during the fabrication of DDS. Physical modification is more reliable and is less likely to alter the original structure of the polysaccharides. However, chemical modifications, which are sometimes performed to obtain polysaccharide derivatives, can disrupt HPS activity. In such cases, the structure–activity relationship of polysaccharides should be taken into consideration, as the active sites of the polysaccharides could change after modification. Specifically, in some cases, the polysaccharide chain can break down after modification, leading to a loss or change in function. Therefore, reasonable and efficient chemical modification strategies are necessary.
Despite the significant efforts made towards understanding the potential of HPS in DDS, several aspects remain to be elucidated. However, given the primary focus on evaluating the influence of different hydrophobic modifications, the effectiveness of different combination modes and sites of polysaccharides and polymers/drugs has received limited attention. Moreover, the structure of HPS–polymer derivatives needs to be evaluated further to understand the related self-assembly mechanism. In summary, to fabricate HPS-based NPs with desirable and controllable properties for biomedical applications, the interactions between HPS and other components should be comprehensively analyzed in future studies.

Funding

This research was funded by the National Natural Science Foundation of China (81760642; 82060645) and the Key Natural Science Foundation of Yunnan Province (202002AA100004-2, 202102AA310045, 202002AA1000056, and 202101AT070099).

Conflicts of Interest

The authors declare no competing financial interests.

Abbreviations

HPS, Herb polysaccharides; DDS, drug delivery systems; HPS-DDS, HPS-based DDS; HA, hyaluronic acid; CS,Chitosan; NK, natural killer; TME, tumor microenvironment; NPs, nanoparticles; APC, antigen presenting cells; DCs, dendritic cells; PLGA, poly(lactic-co-glycolic acid); PLA, poly(D,L-lactide); HPS-MNPs, HPS-based metallic NPs; STMP, sodium trimetaphosphate; TGP, tamarind gum polysaccharides; GPSL, liposomes based on Glycyrrhizophores polysaccharides; OVA, ovalbumin; MP, Lepidium meyenii Walp(maca) polysaccharides; GLP, Gracilaria lemaneiformis polysaccharides; ALPS, Albizia lebbeck L. polysaccharides; VBCP, vinegar baked Radix Bupleurum polysaccharides; RMPs, Ramulus mori polysaccharides; DOTP, Dioscorea opposite Thunb. Polysaccharides; ALPs, Amomum longiligulare polysaccharides; CYP, Chinese yam polysaccharides; PEI, polyethyleneimine; DP, dendrobium polysaccharide; RGPSs, Rehmannia glutinosa polysaccharides; His, histidine; CYS, cystamine; ASGPR, Asialoglycoprotein receptor; MRs, Mannose receptors; TAMs, tumor-associated macrophages; TIDCs, tumor-infiltrating dendritic cells; CQ, chloroquine;ASPs Angelica sinensis polysaccharides; APSs, Astragalus polysaccharides; PSDCs, polysaccharides-drug conjugates; PMPSs, Polygonum multiflorum polysaccharides; Dex, dexamethasone; Dox, doxorubicin; BSPs, Bletilla striata polysaccharides; MNPs, Metallic NPs; CMTs, carboxymethyl tamarind polysaccharides; RP3, Rosa roxburghii Tratt fruit polysaccharides; SPSSs, soluble soybean polysaccharides; MOS-PS, Moringa oleifera seed polysaccharides; PST, Tamarindus indica polysaccharides; LBPs, Lycium barbarum polysaccharides; FFP, Farfarae Flos polysaccharides; DM, diabetes mellitus; MAP, Modified apple polysaccharides; INS, insulin; DPS, Dioscorea opposita Thunb. polysaccharides; Zn, zinc; Se, Selenium; RTFP, Rosa roxburghii fruit polysaccharides; PVA, polyvinyl alcohol; IPNs, interpenetrating networks; MAAc, Psyllium polysaccharides (Psy) and methacrylic acid; DMNs, Dissolving microneedles; PNPS, Panax notoginseng polysaccharides.

References

  1. Muhamad, I.I.; Lazim, N.; Selvakumaran, S.J.N.P.D.D.; Applications, B. Natural polysaccharide-based composites for drug delivery and biomedical applications. ScienceDirect 2019, 419–440. [Google Scholar]
  2. Yu, Y.; Shen, M.; Song, Q.; Xie, J. Biological activities and pharmaceutical applications of polysaccharide from natural resources: A review. Carbohydr. Polym. 2018, 183, 91–101. [Google Scholar] [CrossRef]
  3. Liu, Z.; Jiao, Y.; Wang, Y.; Zhou, C.; Zhang, Z.J.A. Polysaccharides-based nanoparticles as drug delivery systems. Adv. Drug Deliv. Rev. 2008, 60, 1650–1662. [Google Scholar] [CrossRef]
  4. Perumal, P.; Kiruthika, T.; Sivaraj, P.; Lakshmi, D.; Selvin, P.C. Tamarind seed polysaccharide biopolymer-assisted synthesis of spinel zinc iron oxide as a promising alternate anode material for lithium-ion batteries. J. Mater. Sci. Mater. Electron. 2020, 31, 10593–10604. [Google Scholar] [CrossRef]
  5. Abedini, F.; Ebrahimi, M.; Roozbehani, A.H.; Domb, A.J.; Hosseinkhani, H. Overview on natural hydrophilic polysaccharide polymers in drug delivery. Polym. Adv. Technol. 2018, 29, 2564–2573. [Google Scholar] [CrossRef]
  6. Shao, P.; Zhu, Y.; Jin, W. Physical and chemical stabilities of beta-carotene emulsions stabilized by Ulva fasciata polysaccharide. Food Hydrocoll. 2017, 64, 28–35. [Google Scholar] [CrossRef]
  7. Otero, P.; Carpena, M.; Garcia-Oliveira, P.; Echave, J.; Soria-Lopez, A.; Garcia-Perez, P.; Fraga-Corral, M.; Cao, H.; Nie, S.; Xiao, J.; et al. Seaweed polysaccharides: Emerging extraction technologies, chemical modifications and bioactive properties. Crit. Rev. Food Sci. Nutr. 2021, 1–29. [Google Scholar] [CrossRef]
  8. Wang, L.; Wu, Y.; Li, J.; Qiao, H.; Di, L. Rheological and mucoadhesive properties of polysaccharide from Bletilla striata with potential use in pharmaceutics as bio-adhesive excipient. Int. J. Biol. Macromol. 2018, 120, 529–536. [Google Scholar] [CrossRef] [PubMed]
  9. Cha, S.Y.; Park, S.Y.; Lee, J.S.; Lee, K.H.; Kim, J.H.; Fang, Y.; Shin, S.S. Efficacy of Dendrobium candidum polysaccharide extract as a moisturizer. J. Cosmet. Dermatol. 2022, 21, 3117–3126. [Google Scholar] [CrossRef]
  10. Pushpamalar, J.; Veeramachineni, A.K.; Owh, C.; Loh, X.J. Biodegradable Polysaccharides for Controlled Drug Delivery. Chempluschem 2016, 81, 504–514. [Google Scholar] [CrossRef]
  11. Huang, G.; Chen, X.; Huang, H. Chemical Modifications and Biological Activities of Polysaccharides. Curr. Drug Targets 2016, 17, 1799–1803. [Google Scholar] [CrossRef] [PubMed]
  12. Shi, W.; Hass, B.; Kuss, M.A.; Zhang, H.; Ryu, S.; Zhang, D.; Li, T.; Li, Y.-L.; Duan, B. Fabrication of versatile dynamic hyaluronic acid-based hydrogels. Carbohydr. Polym. 2020, 233, 115803. [Google Scholar] [CrossRef]
  13. Azmana, M.; Mahmood, S.; Hilles, A.R.; Rahman, A.; Bin Arifin, M.A.; Ahmed, S. A review on chitosan and chitosan-based bionanocomposites: Promising material for combatting global issues and its applications. Int. J. Biol. Macromol. 2021, 185, 832–848. [Google Scholar] [CrossRef]
  14. Teng, K.; An, Q.; Chen, Y.; Zhang, Y.; Zhao, Y. Recent Development of Alginate-Based Materials and Their Versatile Functions in Biomedicine, Flexible Electronics, and Environmental Uses. Acs. Biomater. Sci. Eng. 2021, 7, 1302–1337. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, J.; Wang, L.; Zhou, H.; Liang, X.-D.; Zhang, M.-T.; Tang, Y.-X.; Wang, J.-H.; Mao, J.-L. The isolation, structural features and biological activities of polysaccharide from Ligusticum chuanxiong: A review. Carbohydr. Polym. 2022, 285, 118971. [Google Scholar] [CrossRef]
  16. Wang, D.; Liu, Y.; Zhao, W. The Adjuvant Effects on Vaccine and the Immunomodulatory Mechanisms of Polysaccharides From Traditional Chinese Medicine. Front. Mol. Biosci. 2021, 8, 655570. [Google Scholar] [CrossRef]
  17. Zeng, Y.; Xiang, Y.; Sheng, R.; Tomas, H.; Rodrigues, J.; Gu, Z.; Zhang, H.; Gong, Q.; Luo, K. Polysaccharide-based nanomedicines for cancer immunotherapy: A review. Bioact. Mater. 2021, 6, 3358–3382. [Google Scholar] [CrossRef] [PubMed]
  18. Wu, W.; Zhang, L.-L.; Zou, J. Research progress on antioxidation effect of traditional Chinese medicine polysaccharides and sports for diabetes prevention and treatment. Zhongguo Zhongyao Zazhi China J. Chin. Mater. Med. 2016, 41, 2591–2599. [Google Scholar]
  19. Shi, C.; Ma, Q.; Ren, M.; Liang, D.; Yu, Q.; Luo, J. Antitumorpharmacological mechanism of the oral liquid of Poriacocos polysaccharide. J. Ethnopharmacol. 2017, 209, 24–31. [Google Scholar] [CrossRef]
  20. Zhao, B.; Zhang, X.; Han, W.; Cheng, J.; Qin, Y. Wound healing effect of an Astragalus membranaceus polysaccharide and its mechanism. Mol. Med. Rep. 2017, 15, 4077–4083. [Google Scholar] [CrossRef] [PubMed]
  21. Lee, D.-Y.; Park, C.W.; Lee, S.J.; Park, H.-R.; Kim, S.H.; Son, S.-U.; Park, J.; Shin, K.-S. Anti-Cancer Effects of Panax ginseng Berry Polysaccharides via Activation of Immune-Related Cells. Front. Pharmacol. 2019, 10, 1411. [Google Scholar] [CrossRef] [PubMed]
  22. Li, C.-X.; Liu, Y.; Zhang, Y.-Z.; Li, J.-C.; Lai, J. Astragalus polysaccharide: A review of its immunomodulatory effect. Arch. Pharmacal. Res. 2022, 45, 367–389. [Google Scholar] [CrossRef]
  23. Wei, W.; Feng, L.; Nie, S.P.; Han, Q.B. Structure characterization and immunomodulating effects of polysaccharides isolated from Dendrobium officinale. Planta Med. 2016, 82, 881–889. [Google Scholar]
  24. Yan, Q.; Long, X.; Zhang, P.; Lei, W.; Sun, D.; Ye, X. Oxidized Bletilla rhizome polysaccharide-based aerogel with synergistic antibiosis and hemostasis for wound healing. Carbohydr. Polym. 2022, 293, 119696. [Google Scholar] [CrossRef]
  25. Sun, Q.; Shi, P.; Lin, C.; Ma, J. Effects of Astragalus Polysaccharides Nanoparticles on Cerebral Thrombosis in SD Rats. Front. Bioeng. Biotechnol. 2020, 8, 616759. [Google Scholar] [CrossRef]
  26. Zhu, J.; Guo, X.; Guo, T.; Yang, Y.; Cui, X.; Pan, J.; Qu, Y.; Wang, C. Novel pH-responsive and self-assembled nanoparticles based on Bletilla striata polysaccharide: Preparation and characterization. Rsc. Adv. 2018, 8, 40308–40320. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, N.; Wardwell, P.R.; Bader, R.A. Polysaccharide-based micelles for drug delivery. Pharmaceutics 2013, 5, 329–352. [Google Scholar] [CrossRef] [PubMed]
  28. Huang, Y.; Liu, Z.; Bo, R.; Xing, J.; Luo, L.; Zhen, S.; Niu, Y.; Hu, Y.; Liu, J.; Wu, Y.; et al. The enhanced immune response of PCV-2 vaccine using Rehmannia glutinosa polysaccharide liposome as an adjuvant. Int. J. Biol. Macromol. 2016, 86, 929–936. [Google Scholar] [CrossRef]
  29. Jiao, L.; Liu, Z.; Zhang, Y.; Feng, Z.; Gu, P.; Huang, Y.; Liu, J.; Wu, Y.; Wang, D. Lentinan PLGA-stabilized pickering emulsion for the enhanced vaccination. Int. J. Pharm. 2022, 611, 121348. [Google Scholar] [CrossRef]
  30. Yang, L.; Han, Z.; Chen, C.; Li, Z.; Yu, S.; Qu, Y.; Zeng, R. Novel probiotic-bound oxidized Bletilla striata polysaccharide-chitosan composite hydrogel. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 117, 121348. [Google Scholar] [CrossRef] [PubMed]
  31. Fonseca, D.F.S.; Vilela, C.; Silvestre, A.J.D.; Freire, C.S.R. A compendium of current developments on polysaccharide and protein-based microneedles. Int. J. Biol. Macromol. 2019, 136, 704–728. [Google Scholar] [CrossRef] [PubMed]
  32. Nai, J.; Zhang, C.; Shao, H.; Li, B.; Li, H.; Gao, L.; Dai, M.; Zhu, L.; Sheng, H. Extraction, structure, pharmacological activities and drug carrier applications of Angelica sinensis polysaccharide. Int. J. Biol. Macromol. 2021, 183, 2337–2353. [Google Scholar] [CrossRef] [PubMed]
  33. Demina, T.S.; Kilyashova, L.A.; Popyrina, T.N.; Svidchenko, E.A.; Bhuniya, S.; Akopova, T.A.; Grandfils, C. Polysaccharides as Stabilizers for Polymeric Microcarriers Fabrication. Polymers 2021, 13, 3045. [Google Scholar] [CrossRef] [PubMed]
  34. Zhao, Y.; Wan, P.; Wang, J.; Li, P.; Hu, Q.; Zhao, R. Polysaccharide from vinegar baked radix bupleuri as efficient solubilizer for water-insoluble drugs of Chinese medicine. Carbohydr. Polym. 2020, 229, 115473. [Google Scholar] [CrossRef] [PubMed]
  35. Tang, Q.; Huang, G. Improving method, properties and application of polysaccharide as emulsifier. Food Chem. 2022, 376, 131937. [Google Scholar] [CrossRef] [PubMed]
  36. Sun, B.; Yu, S.; Zhao, D.; Guo, S.; Wang, X.; Zhao, K. Polysaccharides as vaccine adjuvants. Vaccine 2018, 36, 5226–5234. [Google Scholar] [CrossRef] [PubMed]
  37. Hu, Y.; He, Y.; Niu, Z.; Shen, T.; Zhang, J.; Wang, X.; Hu, W.; Cho, J.Y. A review of the immunomodulatory activities of polysaccharides isolated from Panax species. J. Ginseng Res. 2022, 46, 23–32. [Google Scholar] [CrossRef]
  38. Pang, G.; Wang, F.; Zhang, L.W. Dose matters: Direct killing or immunoregulatory effects of natural polysaccharides in cancer treatment. Carbohydr. Polym. 2018, 195, 243–256. [Google Scholar] [CrossRef] [PubMed]
  39. Mu, M.; Li, X.; Tong, A.; Guo, G. Multi-functional chitosan-based smart hydrogels mediated biomedical application. Expert Opin. Drug Deliv. 2019, 16, 239–250. [Google Scholar] [CrossRef] [PubMed]
  40. Zhu, J.Y.; Tang, X.D.; Jia, Y.; Ho, C.T.; Huang, Q.R. Applications and delivery mechanisms of hyaluronic acid used for topical/transdermal delivery—A review. Int. J. Pharm. 2020, 578, 119127. [Google Scholar] [CrossRef] [PubMed]
  41. Hu, Q.; Lu, Y.; Luo, Y. Recent advances in dextran-based drug delivery systems: From fabrication strategies to applications. Carbohydr. Polym. 2021, 264, 117999. [Google Scholar] [CrossRef] [PubMed]
  42. Niculescu, A.-G.; Grumezescu, A.M. Applications of Chitosan-Alginate-Based Nanoparticles-An Up-to-Date Review. Nanomaterials 2022, 12, 186. [Google Scholar] [CrossRef]
  43. Chen, F.; Huang, G. Preparation and immunological activity of polysaccharides and their derivatives. Int. J. Biol. Macromol. 2018, 112, 211–216. [Google Scholar] [CrossRef] [PubMed]
  44. Xie, J.-H.; Jin, M.-L.; Morris, G.A.; Zha, X.-Q.; Chen, H.-Q.; Yi, Y.; Li, J.-E.; Wang, Z.-J.; Gao, J.; Nie, S.-P.; et al. Advances on Bioactive Polysaccharides from Medicinal Plants. Crit. Rev. Food Sci. Nutr. 2016, 56, S60–S84. [Google Scholar] [CrossRef]
  45. Li, R.; Chen, H.; Zhou, X. Immunomodulatory Effects of Plant Polysaccharides:A Review of the Mechanisms. Nat. Prod. Res. Dev. 2018, 30, 2017. [Google Scholar]
  46. Ren, L.; Zhang, J.; Zhang, T. Immunomodulatory activities of polysaccharides from Ganoderma on immune effector cells. Food Chem. 2021, 340, 127933. [Google Scholar] [CrossRef] [PubMed]
  47. Yi, Y.; Cao, Y.; Zhang, M. Immunomodulatory Activities and Mechanisms of Polysaccharides on T/B Lymphocytes. Chin. J. Cell Biol. 2012, 34, 67–74. [Google Scholar]
  48. Chung, J.T.; Lau, C.M.L.; Chau, Y. The effect of polysaccharide-based hydrogels on the response of antigen-presenting cell lines to immunomodulators. Biomater. Sci. 2021, 9, 6542–6554. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, C.; Liu, S.; Xu, J.; Gao, M.; Qu, Y.; Liu, Y.; Yang, Y.; Cui, X. Dissolvable microneedles based on Panax notoginseng polysaccharide for transdermal drug delivery and skin dendritic cell activation. Carbohydr. Polym. 2021, 268, 118211. [Google Scholar] [CrossRef] [PubMed]
  50. Haogui, X.I.E.; Meizhen, C.; Yuqiang, Z. Research Progress on Structure-antitumor Activity Relationship of Polysaccharide and Its Mechanism. Food Sci. 2011, 32, 329–333. [Google Scholar]
  51. Ma, Z.-X.; Zhao, L.-H. Polysaccharides activate signaling pathways of macrophage. Zhejiang Da Xue Xue Bao Yi Xue Ban J. Zhejiang Univer. Med. Sci. 2011, 40, 567–572. [Google Scholar]
  52. Zhou, X.N.; Dong, Q.; Kan, X.Z.; Peng, L.H.; Xu, X.Y.; Fang, Y.; Yang, J.L. Immunomodulatory activity of a novel polysaccharide from Lonicera japonica in immunosuppressed mice induced by cyclophosphamide. PLoS ONE 2018, 13, e0204152. [Google Scholar] [CrossRef]
  53. Sun, Y.J.; Wang, F.; Liu, Y.; An, Y.Y.; Chang, D.W.; Wang, J.K.; Xia, F.; Liu, N.; Chen, X.F.; Cao, Y.G. Comparison of water- and alkali-extracted polysaccharides from Fuzhuan brick tea and their immunomodulatory effects in vitro and in vivo. Food Funct. 2022, 13, 806–824. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, M.; Dong, L.; Zhang, R. Research Progress on Main Bioactive Substances and Their Health Benefits of Lychee Pulp. J. Food Sci. Technol. 2019, 37, 1–12. [Google Scholar] [CrossRef]
  55. Chen, L.; Huang, G. Antitumor Activity of Polysaccharides: An Overview. Curr. Drug Targets 2018, 19, 89–96. [Google Scholar] [CrossRef]
  56. Guo, R.; Chen, M.; Ding, Y.; Yang, P.; Wang, M.; Zhang, H.; He, Y.; Ma, H. Polysaccharides as Potential Anti-tumor Biomacromolecules—A Review. Front. Nutr. 2022, 9, 838179. [Google Scholar] [CrossRef] [PubMed]
  57. Motshwene, P.G.; Moncrieffe, M.C.; Grossmann, J.G.; Kao, C.; Ayaluru, M.; Sandercock, A.M.; Robinson, C.V.; Latz, E.; Gay, N.J. An Oligomeric Signaling Platform Formed by the Toll-like Receptor Signal Transducers MyD88 and IRAK-4. J. Biol. Chem. 2009, 284, 25404–25411. [Google Scholar] [CrossRef] [PubMed]
  58. Roselaar, S.E.; Daugherty, A. Lipopolysaccharide decreases scavenger receptor mRNA in vivo. J. Interferon Cytokine Res. Off. J. Int. Soc. Interferon Cytokine Res. 1997, 17, 573–579. [Google Scholar] [CrossRef]
  59. Ilchmann, A.; Burgdorf, S.; Scheurer, S.; Waibler, Z.; Nagai, R.; Wellner, A.; Yamamoto, Y.; Yamamoto, H.; Henle, T.; Kurts, C.; et al. Glycation of a food allergen by the Maillard reaction enhances its T-cell immunogenicity: Role of macrophage scavenger receptor class A type I and II. J. Allergy Clin. Immunol. 2010, 125, 175–183. [Google Scholar] [CrossRef] [PubMed]
  60. Sallusto, F.; Cella, M.; Danieli, C.; Lanzavecchia, A. Dendritic cells use macropinocytosis and the mannose receptor to concentrate macromolecules in the major histocompatibility complex class II compartment: Downregulation by cytokines and bacterial products. J. Exp. Med. 1995, 182, 389–400. [Google Scholar] [CrossRef] [PubMed]
  61. Lee, K.Y.; You, H.J.; Jeong, H.G.; Kang, J.S.; Kim, H.M.; Dal Rhee, S.; Jeon, Y.J. Polysaccharide isolated from Poria cocos sclerotium induces NF-kappa B/Rel activation and NOS expression through the activation of p38 kinase in murine macrophages. Int. Immunopharmacol. 2004, 4, 1029–1038. [Google Scholar] [CrossRef]
  62. Li, H.; Xiao, Y.; Li, H.; Jia, Y.; Luo, S.; Zhang, D.; Zhang, L.; Wu, P.; Xiao, C.; Kan, W.; et al. Ganoderma lucidum polysaccharides ameliorated depression-like behaviors in the chronic social defeat stress depression model via modulation of Dectin-1 and the innate immune system. Brain Res. Bull. 2021, 171, 16–24. [Google Scholar] [CrossRef] [PubMed]
  63. Ma, S.; Wang, Q.; Dai, W.; Sun, X.; Chen, C.; Xing, K. Effects of Angelica sinensis polysaccharides on expression of TLR22 pathway-related genes in malabar grouper Epinephelus malabaricus. J. Dalian Ocean Univ. 2017, 32, 33–37. [Google Scholar]
  64. Zhao, H.; Lu, X.; Yue, H.; Liu, Y.; Liu, X.; Huang, X.; Liu, D. Regulation of Astragalus polysaccharide treated colitis mice by JAK/STAT signaling pathway. Chin. Pharmacol. Bull. 2018, 34, 145–146. [Google Scholar]
  65. Shen, C.Y.; Zhang, W.L.; Jiang, J.G. Immune-enhancing activity of polysaccharides from Hibiscus sabdariffa Linn. via MAPK and NF-kappa B signaling pathways in RAW264.7 cells. J. Funct. Foods 2017, 34, 118–129. [Google Scholar] [CrossRef]
  66. Li, K.; Cao, Y.-X.; Jiao, S.-M.; Du, G.-H.; Du, Y.-G.; Qin, X.-M. Structural Characterization and Immune Activity Screening of Polysaccharides with Different Molecular Weights From Astragali Radix. Front. Pharmacol. 2020, 11, 582091. [Google Scholar] [CrossRef]
  67. Zhao, B.; Zhao, J.; Lv, M.; Li, X.; Wang, J.; Yue, Z.; Shi, J.; Zhang, G.; Sui, G. Comparative study of structural properties and biological activities of polysaccharides extracted from Chroogomphus rutilus by four different approaches. Int. J. Biol. Macromol. 2021, 188, 215–225. [Google Scholar] [CrossRef] [PubMed]
  68. Miao, M.; Yu, W.-Q.; Li, Y.; Sun, Y.-L.; Guo, S.-D. Structural Elucidation and Activities of Cordyceps militaris-Derived Polysaccharides: A Review. Front. Nutr. 2022, 9, 898674. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, C.-J.; Guo, J.-Y.; Cheng, H.; Li, L.; Liu, Y.; Shi, Y.; Xu, J.; Yu, H.-T. Spatial structure and anti-fatigue of polysaccharide from Inonotus obliquus. Int. J. Biol. Macromol. 2020, 151, 855–860. [Google Scholar] [CrossRef] [PubMed]
  70. Zhang, X.; Luo, Y.; Wei, G.; Li, Y.; Huang, Y.; Huang, J.; Liu, C.; Huang, R.; Liu, G.; Wei, Z.; et al. Physicochemical and Antioxidant Properties of the Degradations of Polysaccharides from Dendrobium officinale and Their Suitable Molecular Weight Range on Inducing HeLa Cell Apoptosis. Evid. Based Complement. Altern. Med. 2019, 2019, 4127360. [Google Scholar] [CrossRef]
  71. Sun, X.-Y.; Wang, J.-M.; Ouyang, J.-M.; Kuang, L. Antioxidant Activities and Repair Effects on Oxidatively Damaged HK-2 Cells of Tea Polysaccharides with Different Molecular Weights. Oxidative Med. Cell. Longev. 2018, 2018, 1–17. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, S.; Zhuang, X.; Zhang, X.; Han, W.; Liu, Y.; Sun, D.; Guo, W. Enzymatic Modification of Rice Bran Polysaccharides by Enzymes from Grifola Frondosa: Natural Killer Cell Cytotoxicity and Antioxidant Activity. J. Food Sci. 2018, 83, 1948–1955. [Google Scholar] [CrossRef] [PubMed]
  73. Cho, Y.-C.; Park, J.; Cho, S. Anti-Inflammatory and Anti-Oxidative Effects of luteolin-7-O-glucuronide in LPS-Stimulated Murine Macrophages through TAK1 Inhibition and Nrf2 Activation. Int. J. Mol. Sci. 2020, 21, 2007. [Google Scholar] [CrossRef] [PubMed]
  74. Liu, H.; Xu, J.X.; Xu, X.Y.; Yuan, Z.H.; Song, H.; Yang, L.N.; Zhu, D.S. Structure/function relationships of bean polysaccharides: A review. Crit. Rev. Food Sci. Nutr. 2021, 107, 2411–2502. [Google Scholar] [CrossRef] [PubMed]
  75. Yu, M.; Cao, Y.; Xin, G.S.; Ji, C.F.; Gao, S.Y.; Zou, X.; Qi, Z.; Ji, Y.B.; Qiao, A.N.; Ru, X.; et al. Advances in study on structure of polysaccharide. In Proceedings of the International Symposium on Mechanical Engineering and Material Science (ISMEMS), Jeju Island, Korea, 17–19 November 2016; pp. 227–233. [Google Scholar]
  76. Wang, Y.R.; Han, S.W.; Li, R.F.; Cui, B.S.; Ma, X.J.; Qi, X.Z.; Hou, Q.; Lin, M.B.; Bai, J.Y.; Li, S. Structural characterization and immunological activity of polysaccharides from the tuber of Bletilla striata. Int. J. Biol. Macromol. 2019, 122, 628–635. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, B.; Wang, X.; Xiong, Z.; Lu, G.; Ma, W.; Lv, Q.; Wang, L.; Jia, X.; Feng, L. A review on the applications of Traditional Chinese medicine polysaccharides in drug delivery systems. Chin. Med. 2022, 17, 12. [Google Scholar] [CrossRef] [PubMed]
  78. Xu, L.; He, D.; Zhang, C.; Bai, Y.; Zhang, C. The regulate function of polysaccharides and oligosaccharides that with sulfate group on immune-related disease. J. Funct. Foods 2022, 88, 104870. [Google Scholar] [CrossRef]
  79. Xiao, W.; Zhou, P.; Wang, X.; Zhao, R.; Wang, Y. Comparative Characterization and Immunomodulatory Activities of Polysaccharides Extracted from the Radix of Platycodon grandiflorum with Different Extraction Methods. Molecules 2022, 27, 4759. [Google Scholar] [CrossRef]
  80. Wang, Y.; Yang, Z.; Wei, X. Antioxidant activities potential of tea polysaccharide fractions obtained by ultra filtration. Int. J. Biol. Macromol. 2012, 50, 558–564. [Google Scholar] [CrossRef]
  81. Wang, H.; Wei, G.; Liu, F.; Banerjee, G.; Joshi, M.; Bligh, S.W.; Shi, S.; Lian, H.; Fan, H.; Gu, X.; et al. Characterization of two homogalacturonan pectins with immunomodulatory activity from green tea. Int. J. Mol. Sci. 2014, 15, 9963–9978. [Google Scholar] [CrossRef]
  82. Zhang, C.; Gao, F.; Gan, S.; He, Y.A.; Chen, Z.J.; Liu, X.W.; Fu, C.M.; Qu, Y.; Zhang, J.M. Chemical characterization and gastroprotective effect of an isolated polysaccharide fraction from Bletilla striata against ethanol-induced acute gastric ulcer. Food Chem. Toxicol. 2019, 131, 110539. [Google Scholar] [CrossRef]
  83. Liao, Z.C.; Zeng, R.; Hu, L.L.; Maffucci, K.G.; Qu, Y. Polysaccharides from tubers of Bletilla striata: Physicochemical characterization, formulation of buccoadhesive wafers and preliminary study on treating oral ulcer. Int. J. Biol. Macromol. 2019, 122, 1035–1045. [Google Scholar] [CrossRef] [PubMed]
  84. Peng, Q.; Li, M.; Xue, F.; Liu, H.J. Structure and immunobiological activity of a new polysaccharide from Bletilla striata. Carbohydr. Polym. 2014, 107, 119–123. [Google Scholar] [CrossRef]
  85. Jin, M.L.; Zhao, K.; Huang, Q.S.; Xu, C.L.; Shang, P. Isolation, structure and bioactivities of the polysaccharides from Angelica sinensis (Oliv.) Diels: A review. Carbohydr. Polym. 2012, 89, 713–722. [Google Scholar] [CrossRef] [PubMed]
  86. Shunchun, W.; Songshan, S.H.I.; Jian, C.U.I.; Lili, D.; Zhengtao, W.; Zhibi, H.U. Isolation and Structure Analysis of a Hetero-Polysaccharide from Angelica sinensis (Oliv)Diels. Chin. Pharm. J. 2007, 42, 1255–1258. [Google Scholar]
  87. Yang, X.; Zhou, X.; Wang, L.; Yu, X.; Shi, Z.; Ren, G.; Dong, C. Ultrafiltration separation and immune enhancing activity of American ginseng polysaccharide. Sci. Technol. Food Ind. 2014, 35, 49. [Google Scholar]
  88. Wang, L.J.; Yu, X.N.; Yang, X.S.; Li, Y.; Yao, Y.; Lui, E.M.K.; Ren, G.X. Structural and anti-inflammatory characterization of a novel neutral polysaccharide from North American ginseng (Panax quinquefolius). Int. J. Biol. Macromol. 2015, 74, 12–17. [Google Scholar] [CrossRef]
  89. Yan, J.; Wang, Y.; Zhang, X.; Zhao, X.; Ma, J.; Pu, X.; Wang, Y.; Ran, F.; Wang, Y.; Leng, F.; et al. Snakegourd root/Astragalus polysaccharide hydrogel preparation and application in 3D printing. Int. J. Biol. Macromol. 2019, 121, 309–316. [Google Scholar] [CrossRef]
  90. Shi, S.; Lan, X.; Feng, J.; Ni, Y.; Zhu, M.; JingSun; Wang, J. Hydrogel loading 2D montmorillonite exfoliated by anti-inflammatory Lycium barbarum L. polysaccharides for advanced wound dressing. Int. J. Biol. Macromol. 2022, 209 Pt A, 50–58. [Google Scholar] [CrossRef]
  91. Zhao, G.H.; Kan, J.Q.; Li, Z.X.; Chen, Z.D. Structural features and immunological activity of a polysaccharide from Dioscorea opposita Thunb roots. Carbohydr. Polym. 2005, 61, 125–131. [Google Scholar] [CrossRef]
  92. Luque-Alcaraz, A.G.; Lizardi-Mendoza, J.; Goycoolea, F.M.; Higuera-Ciapara, I.; Arguelles-Monal, W. Preparation of chitosan nanoparticles by nanoprecipitation and their ability as a drug nanocarrier. Rsc. Adv. 2016, 6, 59250–59256. [Google Scholar] [CrossRef]
  93. Tang, P.L.; Chew, S.Y.; Hou, X.; Deng, J.; Badri, K. Novel use of sugarcane leaf polysaccharide in kappa-carrageenan blend hydrogel. Biomass Convers. Bioref. 2022. [Google Scholar] [CrossRef]
  94. Xie, L.; Shen, M.; Hong, Y.; Ye, H.; Huang, L.; Xie, J. Chemical modifications of polysaccharides and their anti-tumor activities. Carbohydr. Polym. 2020, 229, 115436. [Google Scholar] [CrossRef]
  95. Li, X.; Weng, Y.; Kong, X.; Zhang, B.; Li, M.; Diao, K.; Zhang, Z.; Wang, X.; Chen, H. A covalently crosslinked polysaccharide hydrogel for potential applications in drug delivery and tissue engineering. J. Mater. Sci. Mater. Med. 2012, 23, 2857–2865. [Google Scholar] [CrossRef] [PubMed]
  96. Fan, Y.; Liu, Y.; Wu, Y.; Dai, F.; Yuan, M.; Wang, F.; Bai, Y.; Deng, H. Natural polysaccharides based self-assembled nanoparticles for biomedical applications—A review. Int. J. Biol. Macromol. 2021, 192, 1240–1255. [Google Scholar] [CrossRef] [PubMed]
  97. Chen, C.; Wang, Y.; Zhang, D.; Wu, X.; Zhao, Y.; Shang, L.; Ren, J.; Zhao, Y. Natural polysaccharide based complex drug delivery system from microfluidic electrospray for wound healing. Appl. Mater. Today 2021, 23, 101000. [Google Scholar] [CrossRef]
  98. Chiesa, E.; Dorati, R.; Pisani, S.; Conti, B.; Bergamini, G.; Modena, T.; Genta, I. The Microfluidic Technique and the Manufacturing of Polysaccharide Nanoparticles. Pharmaceutics 2018, 10, 267. [Google Scholar] [CrossRef] [PubMed]
  99. Akhter, K.F.; Mumin, M.A.; Lui, E.M.K.; Charpentier, P.A. Immunoengineering with Ginseng Polysaccharide Nanobiomaterials through Oral Administration in Mice. Acs. Biomater. Sci. Eng. 2019, 5, 2916–2925. [Google Scholar] [CrossRef]
  100. Zhou, R.; Teng, L.; Zhu, Y.; Zhang, C.; Yang, Y.; Chen, Y. Preparation of Amomum longiligulare polysaccharides 1-PLGA nanoparticle and its immune enhancement ability on RAW264.7 cells. Int. Immunopharmacol. 2021, 99, 108053. [Google Scholar] [CrossRef]
  101. Luo, L.; Zheng, S.; Huang, Y.; Qin, T.; Xing, J.; Niu, Y.; Bo, R.; Liu, Z.; Huang, Y.; Hu, Y.; et al. Preparation and characterization of Chinese yam polysaccharide PLGA nanoparticles and their immunological activity. Int. J. Pharm. 2016, 511, 140–150. [Google Scholar] [CrossRef]
  102. Feng, Z.; Peng, S.; Wu, Z.; Jiao, L.; Xu, S.; Wu, Y.; Liu, Z.; Hu, Y.; Liu, J.; Wu, Y.; et al. Ramulus mori polysaccharide-loaded PLGA nanoparticles and their anti-inflammatory effects in vivo. Int. J. Biol. Macromol. 2021, 182, 2024–2036. [Google Scholar] [CrossRef] [PubMed]
  103. Xu, S.; Wusiman, A.; Liu, Z.; Gu, P.; Ni, H.; Zhang, Y.; Hu, Y.; Liu, J.; Wu, Y.; Wang, D. pH-responsive Astragalus polysaccharides-loaded poly(lactic-co-glycolic acid) nanoparticles and their in vitro immunogenicity. Int. J. Biol. Macromol. 2019, 125, 865–875. [Google Scholar] [CrossRef] [PubMed]
  104. Feng, Z.; Jiao, L.; Wu, Z.; Xu, J.; Gu, P.; Xu, S.; Liu, Z.; Hu, Y.; Liu, J.; Wu, Y.; et al. A Novel Nanomedicine Ameliorates Acute Inflammatory Bowel Disease by Regulating Macrophages and T-Cells. Mol. Pharm. 2021, 18, 3484–3495. [Google Scholar] [CrossRef] [PubMed]
  105. Kumar, S.; Tummala, H. Development of Soluble Inulin Microparticles as a Potent and Safe Vaccine Adjuvant and Delivery System. Mol. Pharm. 2013, 10, 1845–1853. [Google Scholar] [CrossRef] [PubMed]
  106. Luiz, C.; Caires, N.P.; de Aguiar, T.; Lemos Blainski, J.M.; Behs, J.d.S.; Oliveira Coqueiro, D.S.; Di Piero, R.M. Resistance of strawberries to Xanthomonas fragariae induced by aloe polysaccharides and essential oils nanoemulsions is associated with phenolic metabolism and stomata closure. Australas. Plant Pathol. 2022, 51, 305–314. [Google Scholar] [CrossRef]
  107. Ma, F.Y.; Zhang, Y.; Wen, Y.R.; Yao, Y.N.; Zhu, J.H.; Liu, X.H.; Bell, A.; Tikkanen-Kaukanen, C. Emulsification properties of polysaccharides from Dioscorea opposita Thunb. Food Chem. 2017, 221, 919–925. [Google Scholar] [CrossRef]
  108. Gu, P.; Xu, S.; Zhou, S.; Liu, Z.; Sun, Y.; Ou, N.; Hu, Y.; Liu, J.; Wu, Y.; Wang, X.; et al. Optimization of angelica sinensis polysaccharide-loaded Poly (lactic-co-glycolicacid) nanoparticles by RSM and its immunological activity in vitro. Int. J. Biol. Macromol. 2018, 107, 222–229. [Google Scholar] [CrossRef] [PubMed]
  109. Chebil, A.; Funfschilling, D.; Leonard, M.; Six, J.-L.; Nouvel, C.; Durand, A. Amphiphilic Polysaccharides Acting both as Stabilizers and Surface Modifiers during Emulsification in Microfluidic Flow-Focusing Junction. ACS Appl. Bio Mater. 2018, 1, 879–887. [Google Scholar] [CrossRef]
  110. Wang, W.; He, S.; Hong, T.; Zhang, Y.; Sui, H.; Zhang, X.; Ma, Y. Synthesis, self-assembly, and in vitro toxicity of fatty acids-modified Bletilla striata polysaccharide. Artif. Cells Nanomed. Biotechnol. 2017, 45, 69–75. [Google Scholar] [CrossRef] [PubMed]
  111. Zhang, J.; Ji, T.; Yang, X.; Liu, G.; Liang, L.; Liu, X.; Wen, C.; Ye, Z.; Wu, M.; Xu, X. Properties of selenium nanoparticles stabilized by Lycium barbarum polysaccharide-protein conjugates obtained with subcritical water. Int. J. Biol. Macromol. 2022, 205, 672–681. [Google Scholar] [CrossRef] [PubMed]
  112. Beater, E.J.; Onissema-Karimu, S.; Rivera-Galletti, A.; Francis, M.; Witkowski, J.; Salas-de la Cruz, D.; Hu, X. Protein-Polysaccharide Composite Materials: Fabrication and Applications. Polymers 2020, 12, 464. [Google Scholar] [CrossRef]
  113. Zhang, G.; Huang, L.; Wu, J.; Liu, Y.; Zhang, Z.; Guan, Q. Doxorubicin-loaded folate-mediated pH-responsive micelle based on Bletilla striata polysaccharide: Release mechanism, cellular uptake mechanism, distribution, pharmacokinetics, and antitumor effects. Int. J. Biol. Macromol. 2020, 164, 566–577. [Google Scholar] [CrossRef]
  114. Gan, M.; Zhang, W.; Wei, S.; Dang, H. The influence of mPEG-PCL and mPEG-PLGA on encapsulation efficiency and drug-loading of SN-38 NPs. Artif. Cells Nanomed. Biotechnol. 2017, 45, 389–397. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, D.; Wang, L.; Zhang, X.; Bao, D.; Zhao, Y. Polymeric micelles for pH-responsive lutein delivery. J. Drug Deliv. Sci. Technol. 2018, 45, 281–286. [Google Scholar] [CrossRef]
  116. Kim, K.; Lee, C.-S.; Na, K. Light-controlled reactive oxygen species (ROS)-producible polymeric micelles with simultaneous drug-release triggering and endo/lysosomal escape. Chem. Commun. 2016, 52, 2839–2842. [Google Scholar] [CrossRef]
  117. Cao, B.; Zhang, Q.; Guo, J.; Guo, R.; Fan, X.; Bi, Y. Synthesis and evaluation of Grateloupia Livida polysaccharides-functionalized selenium nanoparticles. Int. J. Biol. Macromol. 2021, 191, 832–839. [Google Scholar] [CrossRef] [PubMed]
  118. Luisa Pita-Lopez, M.; Fletes-Vargas, G.; Espinosa-Andrews, H.; Rodriguez-Rodriguez, R. Physically cross-linked chitosan-based hydrogels for tissue engineering applications: A state-of-the-art review. Eur. Polym. J. 2021, 145, 110176. [Google Scholar] [CrossRef]
  119. Matsumoto, Y.; Enomoto, Y.; Kimura, S.; Iwata, T. Highly stretchable curdlan hydrogels and mechanically strong stretched-dried-gel-films obtained by strain-induced crystallization. Carbohydr. Polym. 2021, 269, 118312. [Google Scholar] [CrossRef]
  120. Ma, Y.; Liu, C.; Qu, D.; Chen, Y.; Huang, M.; Liu, Y. Antibacterial evaluation of sliver nanoparticles synthesized by polysaccharides from Astragalus membranaceus roots. Biomed. Pharmacother. 2017, 89, 351–357. [Google Scholar] [CrossRef]
  121. Vokhidova, N.R.; Rashidova, S.S. The influence of synthesis conditions on the film morphology of chitosan-stabilized silver nanoparticles. Polym. Bull. 2022, 79, 3419–3436. [Google Scholar] [CrossRef]
  122. Saini, R.K.; Srivastava, A.K.; Gupta, P.K.; Das, K. pH dependent reversible aggregation of Chitosan and glycol-Chitosan stabilized silver nanoparticles. Chem. Phys. Lett. 2011, 511, 326–330. [Google Scholar] [CrossRef]
  123. Aftab, A.; Bashi, S.; Rafique, S.; Ghani, T.; Khan, R.; Bashir, M.; Ehsan, A.; Khan, M.I.; Shah, A.U.; Mahmood, A. Microfluidic platform for encapsulation of plant extract in chitosan microcarriers embedding silver nanoparticles for breast cancer cells. Appl. Nanosci. 2020, 10, 2281–2293. [Google Scholar] [CrossRef]
  124. Zhang, Y.; Lin, L.; Liu, L.; Liu, F.; Maruyama, A.; Tian, H.; Chen, X. Ionic-crosslinked polysaccharide/PEI/DNA nanoparticles for stabilized gene delivery. Carbohydr. Polym. 2018, 201, 246–256. [Google Scholar] [CrossRef]
  125. Lack, S.; Dulong, V.; Le Cerf, D.; Picton, L.; Argillier, J.F.; Muller, G. Hydrogels based on pullulan crosslinked with sodium trimetaphosphate (STMP): Rheological study. Polym. Bull. 2004, 52, 429–436. [Google Scholar] [CrossRef]
  126. Samrot, A.V.; Angalene, J.L.A.; Roshini, S.M.; Stefi, S.M.; Preethi, R.; Raji, P.; Kumar, A.M.; Paulraj, P.; Kumar, S.S. Purification, characterization and utilization of polysaccharide of Araucaria heterophylla gum for the synthesis of curcumin loaded nanocarrier. Int. J. Biol. Macromol. 2019, 140, 393–400. [Google Scholar] [CrossRef]
  127. Samrot, A.V.; Kudaiyappan, T.; Bisyarah, U.; Mirarmandi, A.; Faradjeva, E.; Abubakar, A.; Selvarani, J.A.; Subbiah, S.K. Extraction, Purification, and Characterization of Polysaccharides of Araucaria heterophylla L and Prosopis chilensis L and Utilization of Polysaccharides in Nanocarrier Synthesis. Int. J. Nanomed. 2020, 15, 7097–7115. [Google Scholar] [CrossRef]
  128. Alavarse, A.C.; Frachini, E.C.G.; da Silva, R.L.C.G.; Lima, V.H.; Shavandi, A.; Petri, D.F.S. Crosslinkers for polysaccharides and proteins: Synthesis conditions, mechanisms, and crosslinking efficiency, a review. Int. J. Biol. Macromol. 2022, 202, 558–596. [Google Scholar] [CrossRef]
  129. Ponrasu, T.; Gu, J.-S.; Wu, J.-J.; Cheng, Y.-S. Evaluation of jelly fig polysaccharide as a shell composite ingredient of colon-specific drug delivery. J. Drug Deliv. Sci. Technol. 2021, 61, 101679. [Google Scholar] [CrossRef]
  130. Cui, X.; Zhang, X.; Yang, Y.; Wang, C.; Zhang, C.; Peng, G. Preparation and evaluation of novel hydrogel based on polysaccharide isolated from Bletilla striata. Pharm. Dev. Technol. 2017, 22, 1001–1011. [Google Scholar] [CrossRef]
  131. 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]
  132. Moniz, T.; Costa Lima, S.A.; Reis, S. Marine polymeric microneedles for transdermal drug delivery. Carbohydr. Polym. 2021, 266, 118098. [Google Scholar] [CrossRef]
  133. Zhuang, X.; Wang, L.; Jiang, X.; Chen, Y.; Zhou, G. The effects of three polysaccharides on the gelation properties of myofibrillar protein: Phase behaviour and moisture stability. Meat Sci. 2020, 170, 108228. [Google Scholar] [CrossRef]
  134. Hyland, L.L.; Taraban, M.B.; Feng, Y.; Hammouda, B.; Yu, Y.B. Viscoelastic properties and nanoscale structures of composite oligopeptide-polysaccharide hydrogels. Biopolymers 2012, 97, 177–188. [Google Scholar] [CrossRef]
  135. Huang, C.; Liao, H.; Ma, X.; Xiao, M.; Liu, X.; Gong, S.; Shu, X.; Zhou, X. Adsorption performance of chitosan Schiff base towards anionic dyes: Electrostatic interaction effects. Chem. Phys. Lett. 2021, 780, 138958. [Google Scholar] [CrossRef]
  136. Lai, M.; Wang, J.; Tan, J.; Luo, J.; Zhang, L.-M.; Deng, D.Y.B.; Yang, L. Preparation, complexation mechanism and properties of nano-complexes of Astragalus polysaccharide and amphiphilic chitosan derivatives. Carbohydr. Polym. 2017, 161, 261–269. [Google Scholar] [CrossRef]
  137. Malviya, R.; Raj, S.; Fuloria, S.; Subramaniyan, V.; Sathasivam, K.; Kumari, U.; Meenakshi, D.U.; Porwal, O.; Kumar, D.H.; Singh, A.; et al. Evaluation of Antitumor Efficacy of Chitosan-Tamarind Gum Polysaccharide Polyelectrolyte Complex Stabilized Nanoparticles of Simvastatin. Int. J. Nanomed. 2021, 16, 2533–2553. [Google Scholar] [CrossRef]
  138. Wang, C.; Luo, W.; Li, P.; Li, S.; Yang, Z.; Hu, Z.; Liu, Y.; Ao, N. Preparation and evaluation of chitosan/alginate porous microspheres/Bletilla striata polysaccharide composite hemostatic sponges. Carbohydr. Polym. 2017, 174, 432–442. [Google Scholar] [CrossRef]
  139. Krystyjan, M.; Khachatryan, G.; Khachatryan, K.; Krzan, M.; Ciesielski, W.; Zarska, S.; Szczepankowska, J. Polysaccharides Composite Materials as Carbon Nanoparticles Carrier. Polymers 2022, 14, 948. [Google Scholar] [CrossRef]
  140. Tabernero, A.; Cardea, S. Microbial Exopolysaccharides as Drug Carriers. Polymers 2020, 12, 2142. [Google Scholar] [CrossRef]
  141. Qian, C.; Zhang, T.; Gravesande, J.; Baysah, C.; Song, X.; Xing, J. Injectable and self-healing polysaccharide-based hydrogel for pH-responsive drug release. Int. J. Biol. Macromol. 2019, 123, 140–148. [Google Scholar] [CrossRef]
  142. Zhou, P.; Zhao, S.; Huang, C.; Qu, Y.; Zhang, C. Bletilla striata polysaccharide microneedle for effective transdermal administration of model protein antigen. Int. J. Biol. Macromol. 2022, 205, 511–519. [Google Scholar] [CrossRef] [PubMed]
  143. Nie, J.; Pei, B.; Wang, Z.; Hu, Q. Construction of ordered structure in polysaccharide hydrogel: A review. Carbohydr. Polym. 2019, 205, 225–235. [Google Scholar] [CrossRef] [PubMed]
  144. Minh-Hiep, N.; Tran, T.-T.; Hadinoto, K. Controlling the burst release of amorphous drug-polysaccharide nanoparticle complex via crosslinking of the polysaccharide chains. Eur. J. Pharm. Biopharm. 2016, 104, 156–163. [Google Scholar]
  145. Monteiro, S.R.; Lopes-da-Silva, J.A. Effect of the molecular weight of a neutral polysaccharide on soy protein gelation. Food Res. Int. 2017, 102, 14–24. [Google Scholar] [CrossRef]
  146. Safronov, A.P.; Terziyan, T.V.; Kyzy, A.M.; Adamova, L.V. Thermodynamic Compatibility of Polyacrylamide with Agarose: The Effect of Polysaccharide Chain Stiffness. Polym. Sci. Ser. A 2022, 64, 53–62. [Google Scholar] [CrossRef]
  147. Usoltseva, R.V.; Shevchenko, N.M.; Malyarenko, O.S.; Ishina, I.A.; Ivannikova, S.I.; Ermakova, S.P. Structure and anticancer activity of native and modified polysaccharides from brown alga Dictyota dichotoma. Carbohydr. Polym. 2018, 180, 21–28. [Google Scholar] [CrossRef]
  148. Wang, X.; Zhao, Y.; Wu, Y.; Liu, L.; Liang, M.; Han, M.; Li, P.; Chen, Z.; Yan, H.; Zhao, R. Size, surface charge and flexibility of vinegar-baked Radix Bupleuri polysaccharide affecting the immune response. Arab. J. Chem. 2022, 15, 8. [Google Scholar] [CrossRef]
  149. Otto, S.; Marina, P.F.; Zhou, F.; Blencowe, A. Thermoresponsive polysaccharides with tunable thermoresponsive properties via functionalisation with alkylamide groups. Carbohydr. Polym. 2021, 254, 117280. [Google Scholar] [CrossRef]
  150. Bruinsmann, F.A.; Pigana, S.; Aguirre, T.; Souto, G.D.; Pereira, G.G.; Bianchera, A.; Fasiolo, L.T.; Colombo, G.; Marques, M.; Pohlmann, A.R.; et al. Chitosan-Coated Nanoparticles: Effect of Chitosan Molecular Weight on Nasal Transmucosal Delivery. Pharmaceutics 2019, 11, 86. [Google Scholar] [CrossRef]
  151. Qi, X.; Wei, W.; Shen, J.; Dong, W. Salecan polysaccharide-based hydrogels and their applications: A review. J. Mater. Chem. B 2019, 7, 2577–2587. [Google Scholar] [CrossRef]
  152. Cui, M.; Zhang, M.; Liu, K. Colon-targeted drug delivery of polysaccharide-based nanocarriers for synergistic treatment of inflammatory bowel disease: A review. Carbohydr. Polym. 2021, 272, 118530. [Google Scholar] [CrossRef] [PubMed]
  153. Nadar, S.S.; Vaidya, L.; Maurya, S.; Rathod, V.K. Polysaccharide based metal organic frameworks (polysaccharide-MOF): A review. Coord. Chem. Rev. 2019, 396, 1–21. [Google Scholar] [CrossRef]
  154. Yang, Y.; Guo, T.; Xu, J.; Xiong, Y.; Cui, X.; Ke, Y.; Wang, C. Micelle nanovehicles for co-delivery of Lepidium meyenii Walp. (maca) polysaccharide and chloroquine to tumor-associated macrophages for synergistic cancer immunotherapy. Int. J. Biol. Macromol. 2021, 189, 577–589. [Google Scholar] [CrossRef] [PubMed]
  155. Wu, Y.; Yi, L.; Li, E.; Li, Y.; Lu, Y.; Wang, P.; Zhou, H.; Liu, J.; Hu, Y.; Wang, D. Optimization of Glycyrrhiza polysaccharide liposome by response surface methodology and its immune activities. Int. J. Biol. Macromol. 2017, 102, 68–75. [Google Scholar] [CrossRef]
  156. Sun, W.; Hu, W.; Meng, K.; Yang, L.; Zhang, W.; Song, X.; Qu, X.; Zhang, Y.; Ma, L.; Fan, Y. Activation of macrophages by the ophiopogon polysaccharide liposome from the root tuber of Ophiopogon japonicus. Int. J. Biol. Macromol. 2016, 91, 918–925. [Google Scholar] [CrossRef]
  157. Shao, P.; Xuan, S.; Wu, W.; Qu, L. Encapsulation efficiency and controlled release of Ganoderma lucidum polysaccharide microcapsules by spray drying using different combinations of wall materials. Int. J. Biol. Macromol. 2019, 125, 962–969. [Google Scholar] [CrossRef]
  158. Wang, J.; Ge, B.; Li, Z.; Guan, F.; Li, F. Structural analysis and immunoregulation activity comparison of five polysaccharides from Angelica sinensis. Carbohydr. Polym. 2016, 140, 6–12. [Google Scholar] [CrossRef]
  159. Xue, L.; Wang, D.; Zhang, D.; Ju, A.; Duan, A.; Xing, J.; Qin, Y.; Yang, S.; Luan, W. The immune adjuvant effect ofAstragaluspolysaccharide onin ovoinjection of Newcastle disease vaccine. J. Anim. Physiol. Anim. Nutr. 2020, 104, 1719–1726. [Google Scholar] [CrossRef]
  160. Liu, C.; Luo, J.; Xue, R.-Y.; Guo, L.; Nie, L.; Li, S.; Ji, L.; Ma, C.-J.; Chen, D.-Q.; Miao, K.; et al. The mucosal adjuvant effect of plant polysaccharides for induction of protective immunity against Helicobacter pylori infection. Vaccine 2019, 37, 1053–1061. [Google Scholar] [CrossRef]
  161. Zhang, Y.; Jiao, L.; Wu, Z.; Gu, P.; Feng, Z.; Xu, S.; Liu, Z.; Yang, Y.; Wang, D. Fabrication and characterization of Chinese yam polysaccharides PLGA nanoparticles stabilized Pickering emulsion as an efficient adjuvant. Int. J. Biol. Macromol. 2022, 209, 513–524. [Google Scholar] [CrossRef]
  162. Zhang, Y.; Cui, Z.; Mei, H.; Xu, J.; Zhou, T.; Cheng, F.; Wang, K. Angelica sinensis polysaccharide nanoparticles as a targeted drug delivery system for enhanced therapy of liver cancer. Carbohydr. Polym. 2019, 219, 143–154. [Google Scholar] [CrossRef] [PubMed]
  163. Lin, W.J.; Lee, W.C. Polysaccharide-modified nanoparticles with intelligent CD44 receptor targeting ability for gene delivery. Int. J. Nanomed. 2018, 13, 3989–4002. [Google Scholar] [CrossRef] [PubMed]
  164. Li, W.J.; Tang, X.-F.; Shuai, X.-X.; Jiang, C.-J.; Liu, X.; Wang, L.-F.; Yao, Y.-F.; Nie, S.-P.; Xie, M.-Y. Mannose Receptor Mediates the Immune Response to Ganoderma atrum Polysaccharides in Macrophages. J. Agric. Food Chem. 2017, 65, 348–357. [Google Scholar] [CrossRef] [PubMed]
  165. Shahbazi, M.-A.; Sedighi, M.; Bauleth-Ramos, T.; Kant, K.; Correia, A.; Poursina, N.; Sarmento, B.; Hirvonen, J.; Santos, H.A. Targeted Reinforcement of Macrophage Reprogramming Toward M2 Polarization by IL-4-Loaded Hyaluronic Acid Particles. Acs. Omega 2018, 3, 18444–18455. [Google Scholar] [CrossRef] [PubMed]
  166. Zhao, M.; Hou, J.; Zheng, S.; Ma, X.; Fu, X.; Hu, S.; Zhao, K.; Xu, W. Peucedanum praeruptorum Dunn polysaccharides regulate macrophage inflammatory response through TLR2/TLR4-mediated MAPK and NF-KB pathways. Biomed. Pharmacother. 2022, 152, 113258. [Google Scholar] [CrossRef]
  167. Monestier, M.; Charbonnier, P.; Gateau, C.; Cuillel, M.; Robert, F.; Lebrun, C.; Mintz, E.; Renaudet, O.; Delangle, P. ASGPR-Mediated Uptake of Multivalent Glycoconjugates for Drug Delivery in Hepatocytes. Chembiochem 2016, 17, 590–594. [Google Scholar] [CrossRef] [PubMed]
  168. Wang, K.; Xu, J.; Liu, Y.; Cui, Z.; He, Z.; Zheng, Z.; Huang, X.; Zhang, Y. Self-assembled Angelica sinensis polysaccharide nanoparticles with an instinctive liver-targeting ability as a drug carrier for acute alcoholic liver damage protection. Int. J. Pharm. 2020, 577, 118996. [Google Scholar] [CrossRef] [PubMed]
  169. Ma, Y.; He, S.; Ma, X.; Hong, T.; Li, Z.; Park, K.; Wang, W. Silymarin-Loaded Nanoparticles Based on Stearic Acid-Modified Bletilla striata Polysaccharide for Hepatic Targeting. Molecules 2016, 21, 265. [Google Scholar] [CrossRef] [PubMed]
  170. Guo, T.; Yang, Y.; Gao, M.; Qu, Y.; Guo, X.; Liu, Y.; Cui, X.; Wang, C. Lepidium meyenii Walpers polysaccharide and its cationic derivative re-educate tumor-associated macrophages for synergistic tumor immunotherapy. Carbohydr. Polym. 2020, 250, 116904. [Google Scholar] [CrossRef] [PubMed]
  171. Jiang, W.; Fu, Y.; Yang, F.; Yang, Y.; Liu, T.; Zheng, W.; Zeng, L.; Chen, T. Gracilaria Iemaneiformis Polysaccharide as Integrin-Targeting Surface Decorator of Selenium Nanoparticles to Achieve Enhanced Anticancer Efficacy. Acs. Appl. Mater. Interfaces 2014, 6, 13738–13748. [Google Scholar] [CrossRef] [PubMed]
  172. Liu, Q.; Chang, X.; Shan, Y.; Fu, F.; Ding, S. Fabrication and characterization of Pickering emulsion gels stabilized by zein/pullulan complex colloidal particles. J. Sci. Food Agric. 2021, 101, 3630–3643. [Google Scholar] [CrossRef] [PubMed]
  173. Liu, F.; Sun, L.; You, G.; Liu, H.; Ren, X.; Wang, M. Effects of Astragalus polysaccharide on the solubility and stability of 15 flavonoids. Int. J. Biol. Macromol. 2020, 143, 873–880. [Google Scholar] [CrossRef] [PubMed]
  174. Zhou, L.; Song, Z.; Zhang, S.; Li, Y.; Xu, J.; Guo, Y. Construction and antitumor activity of selenium nanoparticles decorated with the polysaccharide extracted from Citrus limon (L.) Burm. f. (Rutaceae). Int. J. Biol. Macromol. 2021, 188, 904–913. [Google Scholar] [CrossRef]
  175. Wang, C.; Gao, X.; Chen, Z.; Chen, Y.; Chen, H. Preparation, Characterization and Application of Polysaccharide-Based Metallic Nanoparticles: A Review. Polymers 2017, 9, 689. [Google Scholar] [CrossRef] [PubMed]
  176. Shi, X.-D.; Tian, Y.-Q.; Wu, J.-L.; Wang, S.-Y. Synthesis, characterization, and biological activity of selenium nanoparticles conjugated with polysaccharides. Crit. Rev. Food Sci. Nutr. 2021, 61, 2225–2236. [Google Scholar] [CrossRef]
  177. Banerjee, A.; Halder, U.; Bandopadhyay, R. Preparations and Applications of Polysaccharide Based Green Synthesized Metal Nanoparticles: A State-of-the-Art. J. Clust. Sci. 2017, 28, 1803–1813. [Google Scholar] [CrossRef]
  178. Chen, W.; Cheng, H.; Xia, W. Construction of Polygonatum sibiricum Polysaccharide Functionalized Selenium Nanoparticles for the Enhancement of Stability and Antioxidant Activity. Antioxidants 2022, 11, 240. [Google Scholar] [CrossRef] [PubMed]
  179. Meng, Y.; Zhang, Y.; Jia, N.; Qiao, H.; Zhu, M.; Meng, Q.; Lu, Q.; Zu, Y. Synthesis and evaluation of a novel water-soluble high Se-enriched Astragalus polysaccharide nanoparticles. Int. J. Biol. Macromol. 2018, 118, 1438–1448. [Google Scholar] [CrossRef] [PubMed]
  180. Shao, P.; Feng, J.R.; Sun, P.L.; Xiang, N.; Lu, B.Y.; Qiu, D. Recent advances in improving stability of food emulsion by plant polysaccharides. Food Res. Int. 2020, 137, 109376. [Google Scholar] [CrossRef] [PubMed]
  181. Varma, C.A.K.; Kumar, K.J. Formulation and optimization of pH sensitive drug releasing O/W emulsions using Albizia lebbeck L. seed polysaccharide. Int. J. Biol. Macromol. 2018, 116, 239–246. [Google Scholar] [CrossRef] [PubMed]
  182. Liu, J.; Wang, X.C.; Yong, H.M.; Kan, J.; Jin, C.H. Recent advances in flavonoid-grafted polysaccharides: Synthesis, structural characterization, bioactivities and potential applications. Int. J. Biol. Macromol. 2018, 116, 1011–1025. [Google Scholar] [CrossRef]
  183. Wang, S.; Shao, G.; Yang, J.; Zhao, H.; Qu, D.; Zhang, D.; Zhu, D.; He, Y.; Liu, H. Contribution of soybean polysaccharides in digestion of oil-in-water emulsion-based delivery system in an in vitro gastric environment. Food Sci. Technol. Int. 2020, 26, 444–452. [Google Scholar] [CrossRef]
  184. Sun, Y.; Jia, X.; Yang, R.; Qin, X.; Zhou, X.; Zhang, H.; Kong, W.; Zhang, J.; Wang, J. Deep eutectic solvents boosting solubilization and Se-functionalization of heteropolysaccharide: Multiple hydrogen bonds modulation. Carbohydr. Polym. 2022, 284, 119159. [Google Scholar] [CrossRef] [PubMed]
  185. Akhter, K.F.; Mumin, M.A.; Lui, E.M.K.; Charpentier, P.A. Transdermal nanotherapeutics: Panax quinquefolium polysaccharide nanoparticles attenuate UVB-induced skin cancer. Int. J. Biol. Macromol. 2021, 181, 221–231. [Google Scholar] [CrossRef] [PubMed]
  186. Luo, L.; Qin, T.; Huang, Y.; Zheng, S.; Bo, R.; Liu, Z.; Xing, J.; Hu, Y.; Liu, J.; Wang, D. Exploring the immunopotentiation of Chinese yam polysaccharide poly(lactic-co-glycolic acid) nanoparticles in an ovalbumin vaccine formulation in vivo. Drug Deliv. 2017, 24, 1099–1111. [Google Scholar] [CrossRef] [PubMed]
  187. Gu, P.; Liu, Z.; Sun, Y.; Ou, N.; Hu, Y.; Liu, J.; Wu, Y.; Wang, D. Angelica sinensis polysaccharide encapsulated into PLGA nanoparticles as a vaccine delivery and adjuvant system for ovalbumin to promote immune responses. Int. J. Pharm. 2019, 554, 72–80. [Google Scholar] [CrossRef] [PubMed]
  188. Gu, P.; Wusiman, A.; Zhang, Y.; Cai, G.; Xu, S.; Zhu, S.; Liu, Z.; Hu, Y.; Liu, J.; Wang, D. Polyethylenimine-coated PLGA nanoparticles-encapsulated Angelica sinensis polysaccharide as an adjuvant for H9N2 vaccine to improve immune responses in chickens compared to Alum and oil-based adjuvants. Vet. Microbiol. 2020, 251, 108894. [Google Scholar] [CrossRef] [PubMed]
  189. Zhang, Z.; Li, D.; Li, X.; Guo, Z.; Liu, Y.; Ma, X.; Zheng, S. PEI-modified macrophage cell membrane-coated PLGA nanoparticles encapsulating Dendrobium polysaccharides as a vaccine delivery system for ovalbumin to improve immune responses. Int. J. Biol. Macromol. 2020, 165, 239–248. [Google Scholar] [CrossRef] [PubMed]
  190. Guan, Q.; Sun, D.; Zhang, G.; Sun, C.; Wang, M.; Ji, D.; Yang, W. Docetaxel-Loaded Self-Assembly Stearic Acid-Modified Bletilla striata Polysaccharide Micelles and Their Anticancer Effect: Preparation, Characterization, Cellular Uptake and In Vitro Evaluation. Molecules 2016, 21, 1641. [Google Scholar] [CrossRef]
  191. Guan, Q.; Zhang, G.; Sun, D.; Wang, Y.; Liu, K.; Wang, M.; Sun, C.; Zhang, Z.; Li, B.; Lv, J. In vitro and in vivo evaluation of docetaxel-loaded stearic acid-modified Bletilla striata polysaccharide copolymer micelles. PLoS ONE 2017, 12, e0173172. [Google Scholar] [CrossRef]
  192. Wang, C.; Zhu, J.; Ma, J.; Yang, Y.; Cui, X. Functionalized Bletilla striata polysaccharide micelles for targeted intracellular delivery of Doxorubicin: In vitro and in vivo evaluation. Int. J. Pharm. 2019, 567, 118436. [Google Scholar] [CrossRef]
  193. Liu, Y.; Wu, J.; Huang, L.; Qiao, J.; Wang, N.; Yu, D.; Zhang, G.; Yu, S.; Guan, Q. Synergistic effects of antitumor efficacy via mixed nano-size micelles of multifunctional Bletilla striata polysaccharide-based copolymer and D-alpha-tocopheryl polyethylene glycol succinate. Int. J. Biol. Macromol. 2020, 154, 499–510. [Google Scholar] [CrossRef]
  194. Huang, Z.; Gan, J.; Long, Z.; Guo, G.; Shi, X.; Wang, C.; Zang, Y.; Ding, Z.; Chen, J.; Zhang, J.; et al. Targeted delivery of let-7b to reprogramme tumor-associated macrophages and tumor infiltrating dendritic cells for tumor rejection. Biomaterials 2016, 90, 72–84. [Google Scholar] [CrossRef]
  195. Zhan, X.; Jia, L.; Niu, Y.; Qi, H.; Chen, X.; Zhang, Q.; Zhang, J.; Wang, Y.; Dong, L.; Wang, C. Targeted depletion of tumour-associated macrophages by an alendronate-glucomannan conjugate for cancer immunotherapy. Biomaterials 2014, 35, 10046–10057. [Google Scholar] [CrossRef]
  196. Huang, Y.; Nan, L.; Xiao, C.; Su, F.; Li, K.; Ji, Q.-A.; Wei, Q.; Liu, Y.; Bao, G. PEGylated nano-Rehmannia glutinosa polysaccharide induces potent adaptive immunity against Bordetella bronchiseptica. Int. J. Biol. Macromol. 2021, 168, 507–517. [Google Scholar] [CrossRef]
  197. Pang, G.; Chen, C.; Liu, Y.; Jiang, T.; Yu, H.; Wu, Y.; Wang, Y.; Wang, F.-J.; Liu, Z.; Zhang, L.W. Bioactive Polysaccharide Nanoparticles Improve Radiation-Induced Abscopal Effect through Manipulation of Dendritic Cells. Acs. Appl. Mater. Interfaces 2019, 11, 42661–42670. [Google Scholar] [CrossRef]
  198. Ma, W.; Zhou, Y.; Lou, W.; Wang, B.; Li, B.; Liu, X.; Yang, J.; Yang, B.; Liu, J.; Di, D. Mechanism regulating the inhibition of lung cancer A549 cell proliferation and structural analysis of the polysaccharide Lycium barbarum. Food Biosci. 2022, 47, 101664. [Google Scholar] [CrossRef]
  199. Zhang, Q.; Xu, Y.; Lv, J.; Cheng, M.; Wu, Y.; Cao, K.; Zhang, X.; Mou, X.; Fan, Q. New utilization of Polygonum multiflorum polysaccharide as macromolecular carrier of 5-fluorouracil for controlled release and immunoprotection. Int. J. Biol. Macromol. 2018, 116, 1310–1316. [Google Scholar] [CrossRef]
  200. Wang, L.; Tian, Y.; Zhang, P.; Li, C.; Chen, J. Polysaccharide isolated from Rosa roxburghii Tratt fruit as a stabilizing and reducing agent for the synthesis of silver nanoparticles: Antibacterial and preservative properties. J. Food Meas. Charact. 2022, 16, 1241–1251. [Google Scholar] [CrossRef]
  201. Pang, G.; Zhang, S.; Zhou, X.; Yu, H.; Wu, Y.; Jiang, T.; Zhang, X.; Wang, F.; Wang, Y.; Zhang, L.W. Immunoactive polysaccharide functionalized gold nanocomposites promote dendritic cell stimulation and antitumor effects. Nanomedicine 2019, 14, 1291–1306. [Google Scholar] [CrossRef]
  202. Zhang, Y.; Khan, M.Z.H.; Yuan, T.; Zhang, Y.; Liu, X.; Du, Z.; Zhao, Y. Preparation and characterization of D. opposita Thunb polysaccharide-zinc inclusion complex and evaluation of anti-diabetic activities. Int. J. Biol. Macromol. 2019, 121, 1029–1036. [Google Scholar] [CrossRef]
  203. Taher, M.A.; Khojah, E.; Darwish, M.S.; Elsherbiny, E.A.; Elawady, A.A.; Dawood, D.H. Biosynthesis of Silver Nanoparticles by Polysaccharide of Leucaena leucocephala Seeds and Their Anticancer, Antifungal Properties and as Preservative of Composite Milk Sample. J. Nanomater. 2022, 2022, 7490221. [Google Scholar] [CrossRef]
  204. Devi, I.M.P.; Nallamuthu, N.; Rajini, N.; Kumar, S.M.T.; Siengchin, S.; Rajulu, V.A.; Hariram, N. Antimicrobial properties of poly(propylene) carbonate/Ag nanoparticle-modified tamarind seed polysaccharide with composite films. Ionics 2019, 25, 3461–3471. [Google Scholar]
  205. Sanyasi, S.; Majhi, R.K.; Kumar, S.; Mishra, M.; Ghosh, A.; Suar, M.; Satyam, P.V.; Mohapatra, H.; Goswami, C.; Goswami, L. Polysaccharide-capped silver Nanoparticles inhibit biofilm formation and eliminate multi-drug-resistant bacteria by disrupting bacterial cytoskeleton with reduced cytotoxicity towards mammalian cells. Sci. Rep. 2016, 6, 24929. [Google Scholar] [CrossRef] [PubMed]
  206. Joseph, M.M.; Aravind, S.R.; Varghese, S.; Mini, S.; Sreelekha, T.T. PST-Gold nanoparticle as an effective anticancer agent with immunomodulatory properties. Colloids Surf. B Biointerfaces 2013, 104, 32–39. [Google Scholar] [CrossRef]
  207. Cao, J.; Qin, X.; Li, Z. Synthesis of Silver Nanoparticles from the Polysaccharide of Farfarae Flos and Uncovering Its Anticancer Mechanism Based on the Cell Metabolomic Approach. J. Proteome Res. 2022, 21, 172–181. [Google Scholar] [CrossRef]
  208. Wang, L.; Xie, J.; Huang, T.; Ma, Y.; Wu, Z.Q. Characterization of silver nanoparticles biosynthesized using crude polysaccharides of Psidium guajava L. leaf and their bioactivities. Mater. Lett. 2017, 208, 126–129. [Google Scholar] [CrossRef]
  209. Liu, J.; Ma, Z.X.; Liu, Y.C.; Zheng, X.J.; Pei, Y.; Tang, K.Y. Soluble soybean polysaccharide films containing in-situ generated silver nanoparticles for antibacterial food packaging applications. Food Packag. Shelf Life 2022, 31, 100800. [Google Scholar] [CrossRef]
  210. Mehwish, H.M.; Liu, G.; Rajoka, M.S.R.; Cai, H.; Zhong, J.; Song, X.; Xia, L.; Wang, M.; Aadil, R.M.; Inam-Ur-Raheem, M.; et al. Therapeutic potential of Moringa oleifera seed polysaccharide embedded silver nanoparticles in wound healing. Int. J. Biol. Macromol. 2021, 184, 144–158. [Google Scholar] [CrossRef]
  211. Kumari, Y.; Singh, S.K.; Kumar, R.; Kumar, B.; Kaur, G.; Gulati, M.; Tewari, D.; Gowthamarajan, K.; Karri, V.V.S.N.R.; Ayinkamiye, C.; et al. Modified apple polysaccharide capped gold nanoparticles for oral delivery of insulin. Int. J. Biol. Macromol. 2020, 149, 976–988. [Google Scholar] [CrossRef]
  212. Cai, W.; Hu, T.; Bakry, A.M.; Zheng, Z.; Xiao, Y.; Huang, Q. Effect of ultrasound on size, morphology, stability and antioxidant activity of selenium nanoparticles dispersed by a hyperbranched polysaccharide from Lignosus rhinocerotis. Ultrason. Sonochem. 2018, 42, 823–831. [Google Scholar] [CrossRef] [PubMed]
  213. Zhang, J.; Yang, X.; Ji, T.; Wen, C.; Ye, Z.; Liu, X.; Liang, L.; Liu, G.; Xu, X. Digestion and absorption properties of Lycium barbarum polysaccharides stabilized selenium nanoparticles. Food Chem. 2022, 373, 131637. [Google Scholar] [CrossRef] [PubMed]
  214. Liu, G.; Yang, X.; Zhang, J.; Liang, L.; Miao, F.; Ji, T.; Ye, Z.; Chu, M.; Ren, J.; Xu, X. Synthesis, stability and anti-fatigue activity of selenium nanoparticles stabilized by Lycium barbarum polysaccharides. Int. J. Biol. Macromol. 2021, 179, 418–428. [Google Scholar] [CrossRef] [PubMed]
  215. Zhong, J.-X.; Jin, S.-S.; Wu, K.-S.; Yu, G.-C.; Tu, L.-L.; Liu, L. Effect of nano-selenium loaded with lycium barbarum polysaccharide on the proliferation of lens epithelial cells after UVB damage in vitro. Int. J. Ophthalmol. 2022, 15, 9–14. [Google Scholar] [CrossRef]
  216. Zhang, W.; Zhang, J.; Ding, D.; Zhang, L.; Muehlmann, L.A.; Deng, S.-E.; Wang, X.; Li, W.; Zhang, W. Synthesis and antioxidant properties of Lycium barbarum polysaccharides capped selenium nanoparticles using tea extract. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1463–1470. [Google Scholar] [CrossRef]
  217. Yu, J.; Dong, X.-D.; Jiao, J.-S.; Yu, S.-S.; Ji, H.-Y.; Liu, A.-J.; Chen, Y. The inhibitory effects of selenium nanoparticles modified by fructose-enriched polysaccharide from Codonopsis pilosula on HepG2 cells. Ind. Crops Prod. 2022, 176, 114335. [Google Scholar] [CrossRef]
  218. Zhang, S.; Song, Z.; Shi, L.; Zhou, L.; Zhang, J.; Cui, J.; Li, Y.; Jin, D.-Q.; Ohizumi, Y.; Xu, J.; et al. A dandelion polysaccharide and its selenium nanoparticles: Structure features and evaluation of anti-tumor activity in zebrafish models. Carbohydr. Polym. 2021, 270, 118365. [Google Scholar] [CrossRef]
  219. Xiang, J.; Wang, Y.; Yang, L.; Zhang, X.; Hong, Y.; Shen, L. A novel hydrogel based on Bletilla striata polysaccharide for rapid hemostasis: Synthesis, characterization and evaluation. Int. J. Biol. Macromol. 2022, 196, 1–12. [Google Scholar] [CrossRef]
  220. Ding, L.; Shan, X.; Zhao, X.; Zha, H.; Chen, X.; Wang, J.; Cai, C.; Wang, X.; Li, G.; Hao, J.; et al. Spongy bilayer dressing composed of chitosan-Ag nanoparticles and chitosan-Bletilla striata polysaccharide for wound healing applications. Carbohydr. Polym. 2017, 157, 1538–1547. [Google Scholar] [CrossRef]
  221. Singh, B.; Kumar, S. Synthesis and characterization of psyllium-NVP based drug delivery system through radiation crosslinking polymerization. Nucl. Instrum. Methods Phys. Res. Sect. B-Beam Interact. Mater. At. 2008, 266, 3417–3430. [Google Scholar] [CrossRef]
  222. Preethi, G.U.; Unnikrishnan, B.S.; Sreekutty, J.; Archana, M.G.; Anupama, M.S.; Shiji, R.; Pillai, R.K.; Joseph, M.M.; Syama, H.P.; Sreelekha, T.T. Semi-interpenetrating nanosilver doped polysaccharide hydrogel scaffolds for cutaneous wound healing. Int. J. Biol. Macromol. 2020, 142, 712–723. [Google Scholar] [CrossRef] [PubMed]
  223. Hu, L.; Liao, Z.; Hu, Q.; Maffucci, K.G.; Qu, Y. Novel Bletilla striata polysaccharide microneedles: Fabrication, characterization, and in vitro transcutaneous drug delivery. Int. J. Biol. Macromol. 2018, 117, 928–936. [Google Scholar] [CrossRef] [PubMed]
  224. Zhang, N.; Xue, L.; Younas, A.; Liu, F.; Sun, J.; Dong, Z.; Zhao, Y. Co-delivery of triamcinolone acetonide and verapamil for synergistic treatment of hypertrophic scars via carboxymethyl chitosan and Bletilla striata polysaccharide-based microneedles. Carbohydr. Polym. 2022, 284, 119219. [Google Scholar] [CrossRef] [PubMed]
  225. Feng, Z.; Guan, D.; Zhang, X. Quality standards for the Cordyceps sinensis Sacc polysaccharide liposomal oral liquid. J. Shenyang Pharm. Univ. 2005, 22, 203–206. [Google Scholar]
  226. Bo, R.N.; Zheng, S.S.; Xing, J.; Luo, L.; Niu, Y.L.; Huang, Y.; Liu, Z.G.; Hu, Y.L.; Liu, J.G.; Wu, Y.; et al. The immunological activity of Lycium barbarum polysaccharides liposome in vitro and adjuvanticity against PCV2 in vivo. Int. J. Biol. Macromol. 2016, 85, 294–301. [Google Scholar] [CrossRef]
  227. Akhter, K.F.; Mumin, M.A.; Lui, E.K.; Charpentier, P.A. Microfluidic Synthesis of Ginseng Polysaccharide Nanoparticles for Immunostimulating Action on Macrophage Cell Lines. Acs. Biomater. Sci. Eng. 2016, 2, 96–103. [Google Scholar] [CrossRef]
  228. Akhter, K.F.; Mumin, M.A.; Lui, E.M.K.; Charpentier, P.A. Fabrication of fluorescent labeled ginseng polysaccharide nanoparticles for bioimaging and their immunomodulatory activity on macrophage cell lines. Int. J. Biol. Macromol. 2018, 109, 254–262. [Google Scholar] [CrossRef]
  229. Alalaiwe, A.; Roberts, G.; Carpinone, P.; Munson, J.; Roberts, S. Influence of PEG coating on the oral bioavailability of gold nanoparticles in rats. Drug Deliv. 2017, 24, 591–598. [Google Scholar] [CrossRef]
  230. Moon, H.J.; Lee, S.R.; Shim, S.N.; Jeong, S.H.; Stonik, V.A.; Rasskazov, V.A.; Zvyagintseva, T.; Lee, Y.H. Fucoidan inhibits UVB-induced MMP-1 expression in human skin fibroblasts. Biol. Pharm. Bull. 2008, 31, 284–289. [Google Scholar] [CrossRef]
  231. Lee, H.J.; Kim, J.S.; Song, M.S.; Seo, H.S.; Moon, C.; Kim, J.C.; Jo, S.K.; Jang, J.S.; Kim, S.H. Photoprotective Effect of Red Ginseng against Ultraviolet Radiation-induced Chronic Skin Damage in the Hairless Mouse. Phytother. Res. 2009, 23, 399–403. [Google Scholar] [CrossRef]
  232. Zhang, Y.; Gu, P.; Wusiman, A.; Xu, S.; Ni, H.; Qiu, T.; Liu, Z.; Hu, Y.; Liu, J.; Wang, D. The Immunoenhancement Effects of Polyethylenimine-Modified Chinese Yam Polysaccharide-Encapsulated PLGA Nanoparticles as an Adjuvant. Int. J. Nanomed. 2020, 15, 5527–5543. [Google Scholar] [CrossRef] [PubMed]
  233. Zhang, M.; Sun, L.; Zhao, W.; Peng, X.; Liu, F.; Wang, Y.; Bi, Y.; Zhang, H.; Zhou, Y. Cholesteryl-Modification of a Glucomannan from Bletilla striata and Its Hydrogel Properties. Molecules 2014, 19, 9089–9100. [Google Scholar] [CrossRef]
  234. Liu, Y.; Sun, C.; Zhang, G.; Wu, J.; Huang, L.; Qiao, J.; Guan, Q. Bio-responsive Bletilla striata polysaccharide-based micelles for enhancing intracellular docetaxel delivery. Int. J. Biol. Macromol. 2020, 142, 277–287. [Google Scholar] [CrossRef] [PubMed]
  235. Zijderhand-Bleekemolen, J.E.; Schwartz, A.L.; Slot, J.W.; Strous, G.J.; Geuze, H.J. Ligand- and weak base-induced redistribution of asialoglycoprotein receptors in hepatoma cells. J. Cell Biol. 1987, 104, 1647–1654. [Google Scholar] [CrossRef] [PubMed]
  236. Ekladious, I.; Colson, Y.L.; Grinstaff, M.W. Polymer-drug conjugate therapeutics: Advances, insights and prospects. Nat. Rev. Drug Discov. 2019, 18, 273–294. [Google Scholar] [CrossRef] [PubMed]
  237. Homsi, J.; Simon, G.R.; Garrett, C.R.; Springett, G.; De Conti, R.; Chiappori, A.; Munster, P.N.; Burton, M.K.; Stromatt, S.; Allievi, C.; et al. Phase I trial of poly-L-glutamate camptothecin (CT-2106) administered weekly in patients with advanced solid malignancies. Clin. Cancer Res. 2007, 13, 5855–5861. [Google Scholar] [CrossRef]
  238. Nowotnik, D.P.; Cvitkovic, E. ProLindac (TM) (AP5346): A review of the development of an HPMA DACH platinum Polymer Therapeutic. Adv. Drug Deliv. Rev. 2009, 61, 1214–1219. [Google Scholar] [CrossRef] [PubMed]
  239. Kang, H.; Guan, L.; An, K.; Tian, D. Preparation and physicochemical properties of konjac glucomannan ibuprofen ester as a polysaccharide-drug conjugate. J. Macromol. Sci. Part A-Pure Appl. Chem. 2021, 58, 32–43. [Google Scholar] [CrossRef]
  240. Sauraj; Kumar, S.U.; Gopinath, P.; Negi, Y.S. Synthesis and bio-evaluation of xylan-5-fluorouracil-1-acetic acid conjugates as prodrugs for colon cancer treatment. Carbohydr. Polym. 2017, 157, 1442–1450. [Google Scholar] [CrossRef] [PubMed]
  241. Daus, S.; Heinze, T. Xylan-Based Nanoparticles: Prodrugs for Ibuprofen Release. Macromol. Biosci. 2010, 10, 211–220. [Google Scholar] [CrossRef]
  242. Zhou, S.; Zhang, B.; Liu, X.; Teng, Z.; Huan, M.; Yang, T.; Yang, Z.; Jia, M.; Mei, Q. A New Natural Angelica Polysaccharide Based Colon-Specific Drug Delivery System. J. Pharm. Sci. 2009, 98, 4756–4768. [Google Scholar] [CrossRef] [PubMed]
  243. Prasher, P.; Sharma, M.; Singh, S.P. Drug encapsulating polysaccharide-loaded metal nanoparticles: A perspective drug delivery system. Drug Dev. Res. 2021, 82, 145–148. [Google Scholar] [CrossRef] [PubMed]
  244. Plucinski, A.; Lyu, Z.; Schmidt, B. Polysaccharide nanoparticles: From fabrication to applications. J. Mater. Chem. B 2021, 9, 7030–7062. [Google Scholar] [CrossRef]
  245. Ren, Z.; Luo, Y.; Liu, X.; Zhang, J.; Chen, S.; Yu, R.; Xu, Y.; Meng, Z.; Li, J.; Huang, Y.; et al. Preparation, characterization and controlled-release property of CS crosslinked MWCNT based on Hericium erinaceus polysaccharides. Int. J. Biol. Macromol. 2020, 153, 1310–1318. [Google Scholar] [CrossRef] [PubMed]
  246. Guo, X.; Zhang, J.; Cui, Y.; Chen, S.; Sun, H.; Yang, Q.; Ma, G.; Wang, L.; Kang, J. Highly biocompatible jujube polysaccharide-stabilized palladium nanoparticles with excellent catalytic performance. New J. Chem. 2019, 43, 7646–7652. [Google Scholar] [CrossRef]
  247. Wang, Y.; Han, Q.; Bai, F.; Luo, Q.; Wu, M.; Song, G.; Zhang, H.; Wang, Y. The assembly and antitumor activity of lycium barbarum polysaccharide-platinum-based conjugates. J. Inorg. Biochem. 2020, 205, 111001. [Google Scholar] [CrossRef]
  248. Hu, N.; Gao, Z.; Cao, P.; Song, H.; Hu, J.; Qiu, Z.; Chang, C.; Zheng, G.; Shan, X.; Meng, Y. Uniform and disperse selenium nanoparticles stabilized by inulin fructans from Codonopsis pilosula and their anti-hepatoma activities. Int. J. Biol. Macromol. 2022, 203, 105–115. [Google Scholar] [CrossRef] [PubMed]
  249. Tanaka, Y.; Gong, J.P.; Osada, Y. Novel hydrogels with excellent mechanical performance. Prog. Polym. Sci. 2005, 30, 1–9. [Google Scholar] [CrossRef]
  250. Sahiner, N. Soft and flexible hydrogel templates of different sizes and various functionalities for metal nanoparticle preparation and their use in catalysis. Prog. Polym. Sci. 2013, 38, 1329–1356. [Google Scholar] [CrossRef]
  251. Huseini, H.F.; Rahimzadeh, G.; Fazeli, M.R.; Mehrazma, M.; Salehi, M. Evaluation of wound healing activities of kefir products. Burns 2012, 38, 719–723. [Google Scholar] [CrossRef] [PubMed]
  252. Kumar, K.; Verma, M. Functionalization of psyllium with methacrylic acid through grafting and network formation for use of polymers in water treatment. J. Appl. Polym. Sci. 2007, 103, 1025–1034. [Google Scholar] [CrossRef]
  253. Singh, B. Psyllium as therapeutic and drug delivery agent. Int. J. Pharm. 2007, 334, 1–14. [Google Scholar] [CrossRef] [PubMed]
  254. Chang, C.; Zhang, L. Cellulose-based hydrogels: Present status and application prospects. Carbohydr. Polym. 2011, 84, 40–53. [Google Scholar] [CrossRef]
  255. Damiri, F.; Kommineni, N.; Ebhodaghe, S.O.; Bulusu, R.; Jyothi, V.; Sayed, A.A.; Awaji, A.A.; Germoush, M.O.; Al-Malky, H.S.; Nasrullah, M.Z.; et al. Microneedle-Based Natural Polysaccharide for Drug Delivery Systems (DDS): Progress and Challenges. Pharmaceuticals 2022, 15, 190. [Google Scholar] [CrossRef]
  256. Ohnishi, N.; Yamamoto, E.; Tomida, H.; Hyodo, K.; Ishihara, H.; Kikuchi, H.; Tahara, K.; Takeuchi, H. Rapid determination of the encapsulation efficiency of a liposome formulation using column-switching HPLC. Int. J. Pharm. 2013, 441, 67–74. [Google Scholar] [CrossRef] [PubMed]
  257. Daraee, H.; Etemadi, A.; Kouhi, M.; Alimirzalu, S.; Akbarzadeh, A. Application of liposomes in medicine and drug delivery. Artif. Cells Nanomed. Biotechnol. 2016, 44, 381–391. [Google Scholar] [CrossRef]
  258. Ding, J.; Hu, L.; Kang, X.; Cao, K.; Guan, W. Application of liposome in cancer therapy and tracer study. Chin. J. Clin. Oncol. 2014, 41, 1403–1407. [Google Scholar]
  259. Fan, Y.; Ma, X.; Ma, L.; Zhang, J.; Zhang, W.; Song, X. Antioxidative and immunological activities of ophiopogon polysaccharide liposome from the root of Ophiopogon japonicus. Carbohydr Polym 2016, 135, 110–120. [Google Scholar] [CrossRef]
  260. Wang, S.; Zhu, J.; Cui, X.; Lin, Y.; Yang, Y.; Qu, Y.; Wang, C.; Yang, X. Preparation and evaluation of novel hydrogel of Bletilla striata polysaccharide. Chin. Tradit. Herb. Drugs 2017, 48, 888–893. [Google Scholar]
Figure 1. Fabrication and application of HPS amphiphilic derivatives.
Figure 1. Fabrication and application of HPS amphiphilic derivatives.
Pharmaceutics 14 01703 g001
Figure 2. Fabrication and application of HPS-MNPs. (↑: Increase; ↓: Decrease).
Figure 2. Fabrication and application of HPS-MNPs. (↑: Increase; ↓: Decrease).
Pharmaceutics 14 01703 g002
Figure 3. Fabrication and applications of HPS-based DMNs.
Figure 3. Fabrication and applications of HPS-based DMNs.
Pharmaceutics 14 01703 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cao, Y.; Chen, Z.; Sun, L.; Lin, Y.; Yang, Y.; Cui, X.; Wang, C. Herb Polysaccharide-Based Drug Delivery System: Fabrication, Properties, and Applications for Immunotherapy. Pharmaceutics 2022, 14, 1703. https://doi.org/10.3390/pharmaceutics14081703

AMA Style

Cao Y, Chen Z, Sun L, Lin Y, Yang Y, Cui X, Wang C. Herb Polysaccharide-Based Drug Delivery System: Fabrication, Properties, and Applications for Immunotherapy. Pharmaceutics. 2022; 14(8):1703. https://doi.org/10.3390/pharmaceutics14081703

Chicago/Turabian Style

Cao, Yubiao, Zhuowen Chen, Liangliang Sun, Yameng Lin, Ye Yang, Xiuming Cui, and Chengxiao Wang. 2022. "Herb Polysaccharide-Based Drug Delivery System: Fabrication, Properties, and Applications for Immunotherapy" Pharmaceutics 14, no. 8: 1703. https://doi.org/10.3390/pharmaceutics14081703

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop