Next Article in Journal
Development of Polymersomes Co-Delivering Doxorubicin and Melittin to Overcome Multidrug Resistance
Next Article in Special Issue
Xanthan-Based Materials as a Platform for Heparin Delivery
Previous Article in Journal
Development and Recent Advances in Lysine and N-Terminal Bioconjugation for Peptides and Proteins
Previous Article in Special Issue
All-Polymer Piezo-Composites for Scalable Energy Harvesting and Sensing Devices
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dextran Formulations as Effective Delivery Systems of Therapeutic Agents

by
Anca Roxana Petrovici
1,
Mariana Pinteala
1 and
Natalia Simionescu
1,2,*
1
Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania
2
The Research Institute of the University of Bucharest (ICUB), 90 Sos. Panduri, 050663 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Submission received: 16 December 2022 / Revised: 12 January 2023 / Accepted: 20 January 2023 / Published: 21 January 2023
(This article belongs to the Special Issue New Trends in Polymer-Based Materials)

Abstract

:
Dextran is by far one of the most interesting non-toxic, bio-compatible macromolecules, an exopolysaccharide biosynthesized by lactic acid bacteria. It has been extensively used as a major component in many types of drug-delivery systems (DDS), which can be submitted to the next in-vivo testing stages, and may be proposed for clinical trials or pharmaceutical use approval. An important aspect to consider in order to maintain high DDS’ biocompatibility is the use of dextran obtained by fermentation processes and with a minimum chemical modification degree. By performing chemical modifications, artefacts can appear in the dextran spatial structure that can lead to decreased biocompatibility or even cytotoxicity. The present review aims to systematize DDS depending on the dextran type used and the biologically active compounds transported, in order to obtain desired therapeutic effects. So far, pure dextran and modified dextran such as acetalated, oxidised, carboxymethyl, diethylaminoethyl-dextran and dextran sulphate sodium, were used to develop several DDSs: microspheres, microparticles, nanoparticles, nanodroplets, liposomes, micelles and nanomicelles, hydrogels, films, nanowires, bio-conjugates, medical adhesives and others. The DDS are critically presented by structures, biocompatibility, drugs loaded and therapeutic points of view in order to highlight future therapeutic perspectives.

1. Introduction

Over the last decades, a huge number of macromolecules, including natural polymers, were considered as constituents for drug-delivery systems (DDS) in different formulations: microspheres [1,2], microparticles [3,4], nanoparticles (NPs) [5], nanodroplets [6], liposomes [7], micelles [8,9] and nanomicelles [9], hydrogels [10,11,12], films [13,14], nanowires [15], bio-conjugates [16], medical adhesives [17] and others [18,19,20]. Among natural polymers, polysaccharides are one of the most utilised bio-polymers in DDS’s manufacturing. These compounds are used due to their safety and biocompatibility, the presence of a high variety of chemical functional groups, as well as their high stability and hydrophilic structure. To date, there are a very large number of polysaccharide types isolated and characterised, including dextran (DEX) and its derivatives [3,16], starch and its derivatives [21,22], cellulose and its derivatives [23,24], marine polysaccharides [23], which are used as components in DDS development.
DEX is a noteworthy example of the abovementioned compounds, being a non-toxic, biocompatible, biodegradable and very hydrophilic bio-polymer [25,26]. DEX is biosynthesised intra- or extracellularly by lactic acid bacteria (LAB), which represent one of the most important microbial groups due to their roles in food fermentations and synthesis of techno-functional metabolites [27]. By virtue of its properties, DEX has been used for over 50 years as a circulatory volume expander, in order to improve blood flow [13] and prevent postoperative deep-vein thrombosis [16]. It has also been used in anaemia treatment or as an antiviral agent, being selective for various viruses [13].
In the human body, DEX is degraded by dextranase (1,6-α-D-Glucan 6-glucanohydrolase, E.C. 3.2.1.11) in the liver, spleen, kidney and colon [28,29]. Dextranase endohydrolyses the α-D-(1→6)-glucosidic bonds in DEX resulting in oligosaccharides. The enzyme is synthesised by bacteria present in the colon and after DEX degradation, the by-products are excreted by the kidneys according to the fragments’ molecular weights [30].
In food industries, DEX has technological functions, such as improving the physicochemical properties of food products, and also functional roles, such as prebiotic and immune-modulatory agents [27]. DEX acts as a hydrocolloid in the manufacturing processes of bread and other bakery products, serving as a natural component to replace chemically synthesised commercial hydrocolloids, meeting consumers’ demands for fewer or zero additives in food products. At the same time, it has supplementary properties such as improving dough rheology, textural properties [31] and staling rate [32]. More recently, it was used as a thickener [33], as a surfactant emulsion’s stabiliser [34] and in the production of cereal-based fermented functional beverages and ice cream [35]. The principal potential uses of DEX in foods are mostly related to its capacity to prevent crystallization and retain moisture [36].
In the non-food industry, DEX is used as a bio-separation agent (Sephadex® gels), or as a chromatographic media due to its non-ionic character and good stability under normal operating conditions or for the construction of universal calibration curves used in the evaluation of size exclusion chromatography results [37]. It is used as a steric dispersion stabiliser in the production process of polypyrrole NPs [38].
In the pharmaceutical industry, DEX is already commercially used as a plasma substitute (by increasing volume), as an iron carrier (in the treatment of anaemia, complexed with ferric hydroxide), as an anticoagulant and antithrombotic agent (reducing blood viscosity), as a coating and protective agent for NPs used in nanodrug delivery [25], as an antioxidant and free radical scavenging agent [39], or as inducing agent for interferon biosynthesis [31,35,36,40].
From a medical point of view, the interest in the development and validation of new DDS for different pathologies has grown exponentially. These systems must allow temporal and spatial control of drug delivery, and a continuous plasmatic concentration for a prolonged period and should also improve the drugs’ pharmacokinetic and biopharmaceutical properties. Another very important feature of these systems is that they must provide and increase the drug circulation time and stability in blood flow, improving the drug’s performance, which can be achieved through different types of conjugations with drugs [28].
Over the last decades, DEX has been considered the most promising candidate for the transport of a wide range of therapeutic agents, due to its outstanding physico-chemical properties and biocompatibility [28,41]. Due to the inherent mechanisms of cells which reduce the drug’s effects and facilitate excretion, by using DEX in different DDS, the stability, the local drug concentration and retention time of such nanocarriers (NC) are increased [42].
After systemic administration, the pharmacokinetics of DEX-DDS is considerably influenced by the kinetics of the DEX carrier [41]. Thus, the unmodified polymer can be absorbed by the digestive tract after oral administration only in a small amount. The in vivo studies have shown that both distribution and elimination of DEX depend on the molecular mass and overall charge of the polymer. Pharmacodynamically, the DEX-DDS have resulted in a prolonged effect, a low toxicity profile and a decreased immunogenicity of bioactive molecules [16,43,44].
This review presents a critical and comprehensive overview of the recent developments regarding dextran and its applications for the transport and delivery of drugs, proteins, enzymes, imaging agents, nucleic acids, highlighting the substantial increase in therapeutic potential as compared to the free active principles.

2. DEX Obtained by Biosynthesis from LAB Fermentation

DEX is a polysaccharide which is biosynthesized intra- or extra-cellularly (endopolysaccharide—ENS or exopolysaccharide—EPS) by several microorganisms such as Leuconostoc mesenteroides [31], Leuconostoc dextranicum [45], Lactobacillus brevis, Streptococcus mutants and Weissella confusa [33,35,46], Acetobacter capsulatus, renamed Gluconobacter oxydans and Acetobacter viscous, yeasts and moulds (e.g., Rhizopus spp.) [36]. Commercially, DEX is usually obtained from L. mesenteroides or L. dextranicum fermentation in a media with sucrose and a considerable nitrogen source.
In the biosynthesis of linear polysaccharides, there are two general mechanisms. In the first mechanism, the monomers are sequentially added at the non-reducing end of a growing chain using a high-energy donor. This pathway has been demonstrated for DEX biosynthesized by L. mesenteroides NRRL-B512F [47]. The second mechanism consists of the sequential addition of monomeric units to the reducing end by insertion between a carrier and the growing chain. In both mechanisms, the DEX molecule grows by extrusion, with the enzyme inserting glucose units from sucrose at one end of the polymer chain [36].
The DEX term describes a large class of bacterial extracellular hydrocolloid homo-polysaccharides [37]. DEX is a complex glycan which can be categorised into three types. The first category is represented by DEX with a main chain of consecutive α-D-(1→6)-linked glucose residues with branching at α-D-(1→2), α-D-(1→3), α-D-(1→4). The second DEX type contains non-consecutive α-D-(1→3) and α-D-(1→6) linear linkages and α-D-(1→3) branch linkages, while the third type contains consecutive α-D-(1→6) linear linkages with α-D-(1→6) branch linkages. The configuration of the DEX molecule influences the biopolymer’s water solubility: polymers with predominantly α-D-(1→6) linkages are the most soluble, while DEX with 43% α-D-(1→3) branch linkages are water insoluble. Moreover, DEX is stable in water, dimethyl sulfoxide, formamide, glycerol, 4-methyl morpholine oxide and hexamethyl phosphamide [36].
An important aspect of obtaining high amounts of bio-polymers is the fermentation conditions. Depending on the composition of the culture medium and the strain type, DEX can be obtained with a low or high molecular weight (over 150 kDa) [35,46]. Dextransucrase (1,6-α-D-glucan 6-α-glucosyltransferase, E.C. 2.4.1.5) is a generic name for a family of enzymes that synthesize DEX from sucrose [48]. The activity of dextransucrase is higher in aerobic compared to anaerobic conditions, and the biosynthesis rate are considerably improved by air-sparging [49]. Under proper aeration conditions, sucrose is converted to DEX with maximum yield. Dextransucrase has maximum stability and activity at a pH between 5.0 and 5.5, although most of the published research reports a fermentation pH of around 6.7. At pH 5.5, sucrose is converted into DEX from the beginning of the fermentation process, increasing the conversion yield by approximately 10% in a short period of time [49], preferably in the presence of small amounts of calcium [32]. The optimal biosynthesis temperature range is between 30–45 °C. The enzyme’s nature influences the branching degree of DEX, resulting in different structures of the macromolecule [37]. The molecular weight of biosynthesized DEX is inversely correlated with the dextransucrase concentration and directly correlated with sucrose concentration and temperature [50]. Actually, the dextransucrase cleaves the glycoside bond in sucrose, releasing glucose which is further used in the biosynthesis of DEX by natural polymerisation, and fructose which is used as an energy source in different metabolic processes [51].
To increase the EPS biosynthesized amount, research groups generally optimise the culture media composition by supplementing it with additional carbon and nitrogen sources [52]. Han et al. (2014) [31] obtained 32 g/L DEX from L. mesenteroides BD1710 fermentation in culture media containing tomato juice supplemented with 15% sucrose. Another considerable amount of DEX, about 25.2 g/L, was obtained in our laboratory by W. confusa PP29 fermentation in culture media containing UHT milk supplemented with 8% sucrose [35]. This compound had a remarkable disrupting effect on the biofilm produced by Candida albicans SC5314 strain, as well as no cytotoxic effect on normal human dermal fibroblasts (NHDF) [35]. Wang et al. (2022) [53] simultaneously obtained DEX and vitamin B12 by using Propionibacterium freudenreichii DSM 20,271 and Weissella confusa A1 in a soya flour- or rice bran-based media supplemented with sucrose. The aim of the study was to obtain bread with high nutritional value and the results also showed that the obtained DEX amount was very high, at approximately 58 g/L [53]. Experiments performed in our laboratory showed that the addition of aqueous fruit extract from Hippophae rhamnoides to the LAB culture media yielded 4.8 g/L dry EPS, with 2 g/L more compared with standard MRS media [54], while the addition of anthocyanin-rich Hibiscus sabdariffa L. extracts to culture media supplemented with peptone and sucrose yielded biosynthesized DEX with high molecular weights [55] (see Table 1).

3. Biomedical Applications of Modified DEX

After thorough investigations, different research groups postulated that pure DEX-based systems cannot achieve good mechanical properties and high drug-loading capacity. Native DEX exhibits low-cell-adhesive properties and in order to obtain hydrogels with controlled cell-scaffold interactions, specific molecules must be incorporated [19]. Many research groups have chemically modified DEX by introducing functional groups into the molecule through cross-linking reactions, therefore improving mechanical strength and drug-loading ability [9,41] and increasing the number of compound classes that can be obtained. Furthermore, DEX has been shown to have metal chelating activity [46] and antioxidant properties [59], as well as antitumour activity by regulating apoptosis and autophagy [61].
Below we present the most commonly used types of modified DEX, as well as the active substances that have been loaded into DEX-based systems.

3.1. Acetalated Dextran (Ac-DEX)

The main reason for performing DEX acetylation is to allow solubility of DEX molecules in organic solvents, facilitating the encapsulation of various hydrophilic and hydrophobic active substances, which has always been challenging, and allowing their simultaneous delivery [62]. Ac-DEX is an essential derivative of DEX synthesized in mild conditions, at room temperature, from DEX and 2-methoxypropene in a one-step reaction catalysed by pyridinium p-toluene sulfonate [3]. Ac-DEX contains cyclic and methoxy acyclic acetal moieties and has been shown to be biodegradable at neutral pH, biocompatible and pH-sensitive [4,62]. Because it is an acid-sensitive polymer, Ac-DEX degrades more rapidly at lower pH, for example in the endosome of phagocytic cells, tumours, or in areas with inflammation [63], making it an ideal carrier for a wide range of therapeutics. Ac-DEX has several characteristics that make it a unique biodegradable polymer, such as facile synthesis and degradation rates’ adjustment properties. It is suitable for vaccine applications, targeted host-directed therapies to macrophages, controlled release of drugs, chemotherapeutic delivery and engineered drug-delivery devices [64]. By the simultaneous release of different active substances, synergistic effects, as well as the reduction in side effects and solubility improvement could be achieved at lower concentrations and improved pharmacokinetics [62].
As a therapeutic system, Ac-DEX was used to develop porous microparticles made by single emulsion method in water/oil and loaded with rapamycin [4,65], camptothecin [66], or curcumin [67] in order to be used for pulmonary drug delivery or phagocytes’ passive targeting. The delivery and release tests recorded very good results. These systems are more efficient in drugs’ transport to the alveolar region of the lung, or for immune suppression therapies than other similar systems [4,65,66,67]. At the pulmonary level, after the post-processing of these microparticles, the respirable fraction increased with the improvement of aerosolization and no significant damage was caused by the system to lung epithelial cells either in liquid- or air-exposed conditions [4,65,66,67]. The dry powder aerosol formulations were capable of deep lung delivery of drugs by targeting and releasing the therapeutics to a desired location [4,65,66,67]. By using these systems, a rapid onset of pharmaceutical action was obtained, avoiding hepatic metabolism and decreasing the side effects of the drugs. Resiquimod, a drug with antiviral and antitumour activity, was encapsulated in an electrospun Ac-DEX microparticles’ scaffold and the results were remarkable for tissue engineering, wound healing, immunotherapy and drug-delivery applications [68,69]. Pyraclostrobin, an antifungal agent, was successfully loaded in pH-sensitive Ac-DEX microparticles in order to treat Sclerotinia sclerotiorum plant infections [3]. Konhäuser et al. (2022) [62] developed a DDS system in order to simultaneously release L-asparaginase and etoposide. The active substances have synergistic activity against chronic myeloid leukaemia (CML) K562 cells, but L-asparaginase is hydrophilic and etoposide is hydrophobic [62]. This system has great potential for CML therapy due to its ingenious ability to release both compounds in a pH-dependent manner, leading to synergistic cytotoxicity, increased drug efficacy and reduced side effects [62].

3.2. Oxidized Dextran (oDEX)

Some research groups have obtained oDEX in order to bind therapeutic active molecules for secure delivery. DEX oxidation using sodium periodate is a catalysis-free aqueous reaction which produces a polyaldehydic DEX that can serve as a macromolecular cross-linker for amino groups-bearing substances.
By using oDEX, different DDS were synthesized, including microspheres, vesicles, hydrogels, NPs. Cortesi et al. (1999) [1] synthesized oDEX gelatine microspheres loaded with TAPP-Br antitumour drug and cromoglycate, obtaining very good results for drug release. Curcio et al. (2020) [70] developed a self-assembling oDEX-based vesicular system loaded with camptothecin, which was determined to be very efficient against MCF-7 and MCF-10A cell lines. The antitumour drugs, such as 5-fluorouracil and methotrexate, were encapsulated in oDEX hydrogels for breast, skin and gastrointestinal tract cancer treatment [71]. The obtained DDS induced faster drug release and had excellent biocompatibility and degradability, therefore being suitable for anticancer therapies [71]. Novel oDEX-based NPs for insulin release [29] or loaded with 5-fluorouracil for colorectal cancer therapies [30] were also obtained and were suitable for further in vivo testing.
Zhou et al. (2022) [12] reported an oDEX-based hydrogel loaded with black phosphorus nanosheets and zinc oxide nanoparticles. This DDS was suggested to be a hopeful approach for chronic wound treatment with bacterial infection through the synergistic effect of photothermal action and immunomodulation [12]. Multiple hydrogels as transdermal DDS loaded with ceftazidime or with collagen and Epidermal Growth Factor were reported for the treatment and healing of diabetic wounds infected with multidrug-resistant bacteria [39,72].

3.3. Carboxymethyl Dextran (CMD)

CMD, a polyanionic polysaccharide, was considered as a DDS constituent since it was discovered that its functional groups facilitate chemical conjugation and ionic complexation with various drugs. Its hydrophilic characteristics facilitate prolonged drug circulation improving its tumour-targeting efficiency [73]. By itself, CMD has high antioxidant properties [74].
CMD was used as a nanocomposite hydrophilic shell in order to be loaded with glutathione as an inhibitor of reactive oxygen species’ cytotoxic effects associated with tumour apoptosis [75].
Magnetic NPs were coated with CMD in order to be used as contrast agents for magnetic resonance molecular imaging (MRI) [76,77]. Several research groups used CMD-coated magnetic NPs loaded with antibodies [78], peptides [79] and enzymes [80] for different medical applications.

3.4. Dextran Sulphate Sodium (DSS)

Certain types of dextran functionalization can lead to very toxic compounds, which can, however, be useful for particular applications. DSS is a polyanionic derivative of dextran with high-water solubility properties containing approximately 17% sulphur with up to three sulphate groups (-OSO3Na) per glucose molecule [81]. DSS has found wide utilization in the food, biotechnology, cosmetic and pharmaceutical industries [82]. In proper concentrations, it exhibits positive effects as an anticoagulant and antiviral agent or has the properties of lowering blood lipid and glucose levels in clinical studies [83]. Despite DSS promising application prospects and biological properties, its application is limited due to its harmful effects on the gastrointestinal tract [83].
Different research groups use DSS to induce colitis, thus creating artificial conditions for studying inflammatory bowel diseases, such as ulcerative colitis and Crohn’s disease. The colitogenic potential of DSS depends on its molecular weight which must be between 36–50 kDa. DSS produces manifestations associated with inflammatory bowel disease, such as submucosal erosions, ulceration, inflammatory cell infiltration, crypt abscesses, as well as epithelioglandular hyperplasia [81]. It also determines the shrinkage of colon length and increases the relative colon weight/length ratio accompanied by mucosal oedema and bloody stools [81]. The DSS colitis paradigm is the most appropriate model for the human phenotype, from many points of view. For this injury, many drugs were tested as treatment, including curcumin [84], garlic oil (which has antioxidant, anti-inflammatory and immunomodulatory effects) [85], carvacrol (a phenolic monoterpene extracted from Oreganum vulgarea sp. essential oils with antioxidant, anti-inflammatory and anticancer properties) [86], resveratrol [87], glucose-lysine Maillard reaction products [88], liquorice (a Glycyrrhiza uralensis rhizome-derived product with anti-inflammatory activity) [89], Lactobacillus sakei K040706 (with immuno-stimulatory effects) [90] and Polygonum tinctorium leaves extract (by enhancing the mRNA expression of interleukin-10 and decreasing expression of tumour necrosis factor in colon tissues) [91].
DSS has also been used for film coatings with biological and biomedical applications [13]. Mixed DSS-based systems were developed, such as eco-friendly PVA/DSS nanofibers loaded with ciprofloxacin [18] or chitosan-DSS microparticles loaded with a hydrophilic peptide used as immunity-enhancing adjuvant or considered as vaccine electuary [92].
An antibacterial biocapsule system obtained from multilayer self-assembled diethylaminoethyl (DEAE)-DEX hydrochloride and DSS was developed as a DDS for kanamycin-resistant Escherichia coli treatment. The system manifested an inhibitory effect during bacterial growth having high potential as an antimicrobial agent in future treatments against infection [20].
Wang et al. (2020) [93] developed a dual DDS for paclitaxel and 5-fluorouracil. The pH-sensitive system exhibited a controlled release profile based on a mechanism following a two-phase kinetic model [93]. The system’s efficiency was investigated on HepG2 cells, resulting in synergistic effects between the two drugs and enhanced inhibition of cancer cells, presenting a good potential for biomedical delivery applications [93].

3.5. Diethylaminoethyl-Dextran (DEAE-DEX)

DEAE-DEX was the very first chemical vector used for DNA delivery, reported by Vaheri and Pagano in 1965 as DEAE-DEX used to enhance the cells’ viral infectivity. The DEAE-DEX-mediated transfection method gained attention in the early 1980s because of the simplicity, efficiency and reproducibility of the procedure. DEAE-DEX forms electrostatic interaction complexes with DNA, exhibiting higher transfection efficiency, but at high concentrations, it is toxic to cells [94]. Recently, it was used to develop carrier polyplex nanoparticles with luciferase coding mRNA [95] or used for β-interferon production enhancement [40].

4. Dextran Used in Drug-Delivery Systems

From a structural point of view, as a bio-polymer, DEX has molecular weights higher than 1000 Dalton, and a linear backbone of α-linked D-glucopyranosyl repeating units [28]. DEX contains a large number of hydroxyl groups which are capable of conjugating bioactive molecules by direct coupling or via a linker. DEX has been used to form hydrogels [10,11,12], films [13,96], nanosystems (by itself or as a coating agent) [5,6,9,15,16] and other systems [7,8,17,18,19,20], in order to release controllable amounts of drugs (Table 2). Recently, it was demonstrated that DEX has a protective effect on cells against oxidative stress induced by drug cytotoxicity [28,42].
It has been postulated that in vivo drug concentrations need to be as constant as possible and optimally targeted to specific cells or organs in order to avoid prolonged treatments. Microencapsulation of antineoplastic drugs has been done using natural or synthetic polymeric materials with the aim of maintaining constant and high drug levels in the blood or at the tumour site, thus reducing multiple administrations and possibly targeting the active agents to the desired location [1].
Below, the most used systems containing DEX as a component have been reviewed.

4.1. DEX as a Hydrogel Component

The use of natural polymers in hydrogel systems’ development can confer highly beneficial properties to drugs. By using DEX, optimal release profiles and desirable therapeutic characteristics can be achieved for a wide range of DDS [28]. Hydrogels as polymeric networks with swelling capacity can be biodegradable or not, and drugs can be encapsulated in these structures, obtaining delivery systems with controlled drug release [97].
DEX-containing hydrogels are considered valuable and sustainable biomaterials for biomedical applications [10]. They are being used extensively in the pharmaceutical and biomedical fields for drug delivery, tissue engineering [10], neovascularization [106], regenerative medicine, wound repair and dressings [12,41,107], due to DEX’s lubrification and unique soft-wet properties similar to natural extracellular matrices [108], as well as their advantages for commercial production, such as high yields and low costs [35] (Table 2).
Traditional antibacterial hydrogels deliver large dosages of antibiotics or other drugs, increasing the risk for cytotoxicity. However, some research groups have used antimicrobial agents with synergistic activity in models of normal and diabetic wounds infected with multidrug-resistant bacteria, achieving higher therapeutic effects at lower doses compared to classical antibiotics [72].

4.2. Dextran as NP Component or Coating Agent

Over the years, intensive efforts have been made to design intelligent systems that are able to deliver drugs more efficiently to the target site and at the same time to minimise the side effects. NPs as DDS for enhancing the drugs’ therapeutic efficiency are the hot spot of research in the field of nano-biotechnology. Although there are many advantages associated with these NPs, such as increased solubility of hydrophobic drugs favouring long circulation times in the blood or higher bioavailability [109,110], there are still a number of drawbacks, such as burst release, limited stability of formulations leading to drug leakage and nonspecific cellular uptake resulting in undesired adverse effects [9,44]. Most NPs can be tailored for specific site targeting, controlled release of drugs and high stability under different administration routes. NPs have the ability to penetrate easily through fine blood capillaries due to their subcellular and nano sizes [29,111]. Furthermore, drugs have often been covalently bonded to natural or synthetic polymers in order to reduce renal excretion [109].
DEX in its native form does not self-assemble into NPs, but nonetheless has high water retention capacity and heavy metal chelating activity for Zn2+, Fe2+, Cu2+, Cd2+ and Pb2+ [46]. Different strategies have been developed in order to fabricate DEX-based NPs for drug delivery (Table 3), among which we can mention the covalent functionalization of DEX hydroxyl groups or crosslinking of DEX through the lateral hydroxyl groups (using a variety of crosslinking reactions and linkers), both necessary for physical self-assembly into NPs [112] or reducing in vivo accumulation and clinical risk [30,96,113,114].
In order to safely deliver a drug and to release the correct dose, first of all, it is mandatory to study the physico-chemical properties of the administered drug in the location of interest. Furthermore, in order to selectively target a specific site, it is imperative to investigate the physiological properties of the microenvironment. The toxicity and the bio-distribution of a delivery system are influenced by the chemical nature of the components, system’s size and the coating agents [125]. By using DEX as a coating agent for any NPs, the interactions with cells and proteins are limited, thus conferring increased circulating half-life and colloidal stability in biological environments, which in turn determines good overall safety in vivo and no visible tissular damage [96,129]. At the same time, by the encapsulation of the drug in these systems, the side-effects of the drug are minimized, the efficiency is enhanced and the drug can be released in a controlled rate depending on the drug’s diffusion coefficient [44,71,120,124].

4.3. Dextran as Nanocarrier Component

Nanocarriers (NC) are similar to NPs, but the methods of synthesis are different. Thus, reaction components represented by natural polymers with low molecular weights and various molecules with smaller or larger molecular weights are embedded by chemical or physical processes [44,130]. Next, the final synthesised compound self-assembles through hydrogen interactions or electrostatic attractions in a NC system. Natural or synthetic hydrophobic substances with therapeutic activity are encapsulated either in the core or grafted on the NC surface by chemical reactions or by electrostatic interactions [131].
Similar to NPs, NCs also help improve drug efficacy, having the ability to increase drug absorption in tissue and increase cellular uptake, to protect the drug from degradation and interaction with the biological environment and to control the drug’s pharmacokinetic distribution profile [132]. NCs such as liposomes, micelles or polymeric NPs have shown fabulous opportunities in the field of targeted drug delivery for cancer therapy [133]. Table 4 presents DEX-based NCs developed for drug delivery.

4.4. Dextran as Micelles’ Component

Micelles are a type of highly regarded DDS, especially for the delivery of hydrophobic/lipophilic drugs due to their unique physicochemical properties, containing a hydrophobic core and a hydrophilic shell. Natural polymeric micelles are more widely used in novel DDS due to their biocompatibility and tunable properties [8]. These DDS have a great capacity to encapsulate high amounts of bioactive compounds and to deliver them at targeted locations in the body.
Several groups have developed DEX-based micelles for drug delivery in a variety of pathologies. Zhang et al. (2020) [137] developed a self-assembled pH-responsible micelle formed by conjugated DEX loaded with doxorubicin and found that the drug accumulation in tumours was increased due to permeation enhancement. Jin et al. (2017) [138] tested the cytotoxicity and antitumour activity of their system on MCF-7 and SKOV-3 tumour cells in vitro and the results were promising. Later, a self-assembled DEX-based micelle was loaded with rapamycin, decreasing the drug’s toxicity and increasing the system’s uptake by tumoral cells, without affecting normal cells’ viability [9]. Malekhosseini et al. (2020) synthesized DEX-based micelles which had a hydrocortisone encapsulation efficiency of 79% and 90% drug release in the first 12 h with cell viability higher than 90% [8]. The study of nateglinide and insulin, vitamin E succinate and insulin combinations loaded into DEX-based micelles reduced oxidative stress and improved the mitochondrial function and glucose metabolism, while also improving the cognitive capacity of mice, demonstrating a paradigm for specific and high-efficacy combination therapy for Alzheimer’s disease [139].

5. Conclusions

Dextran is a biosynthesized non-toxic, biocompatible and biodegradable macromolecule which has been extensively used as a major component in many types of DDS due to its versatile properties. Numerous DDS obtained so far using dextran have great potential in different pharmaceutical applications but, in order to maintain the high DDS biocompatibility, the use of dextran obtained by fermentation with minimum chemical modifications is recommended. By performing dextran chemical modifications, artefacts can appear in the DEX spatial structure which can further lead to biocompatibility decreasing or even cytotoxicity increasing. As a result, many DDS containing acetalated, carboxymethyl, diethylaminoethyl-dextran, or dextran sulphate sodium salt have been removed from in vivo or clinical studies.
On the other hand, the multitude of developed DDS (microspheres, microparticles, nanoparticles, nanodroplets, liposomes, micelles, hydrogels, films, nanowires, bio-conjugates, medical adhesives and others) have considerably increased the type and number of applications compatible with DEX-DDS. However, there is still a need for continuous DDS development in order to optimize and study as many systems as possible for biomedical and pharmaceutical applications.

Author Contributions

Conceptualization, A.R.P. and N.S.; resources, A.R.P., M.P. and N.S.; data curation, A.R.P. and N.S.; writing—original draft preparation, A.R.P.; writing—review and editing, M.P. and N.S.; visualization, A.R.P. and N.S.; supervision, M.P.; project administration, M.P.; funding acquisition, M.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant of the Ministry of Research, Innovation and Digitization, CCCDI—UEFISCDI, project number PN-III-P2-2.1-PED-2021-2193, within PNCDI III.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We acknowledge the support provided by the ICUB Fellowship for Young Researchers (N.S., Contract no. 26169/29 November 2022).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cortesi, R.; Esposito, E.; Osti, M.; Squarzoni, G.; Menegatti, E.; Davis, S.S.; Nastruzzi, C. Dextran cross-linked gelatin microspheres as a drug delivery system. Eur. J. Pharm. Biopharm. 1999, 47, 153–160. [Google Scholar] [CrossRef] [PubMed]
  2. Constantin, M.; Bucatariu, S.; Harabagiu, V.; Ascenzi, P.; Fundueanu, G. Do cyclodextrins bound to dextran microspheres act as sustained delivery systems of drugs? Int. J. Pharm. 2014, 469, 1–9. [Google Scholar] [CrossRef] [PubMed]
  3. Xie, Z.; Liang, W.; Xiong, Q.; Zhao, Y.; Cheng, J.; Li, X.; Zhao, J. Acetalated dextran microparticles for the smart delivery of pyraclostrobin to control Sclerotinia diseases. Carbohydr. Polym. 2022, 291, 119576. [Google Scholar] [CrossRef] [PubMed]
  4. Kauffman, K.J.; Kanthamneni, N.; Meenach, S.A.; Pierson, B.C.; Bachelder, E.M.; Ainslie, K.M. Optimization of rapamycin-loaded acetalated dextran microparticles for immunosuppression. Int. J. Pharm. 2012, 422, 356–363. [Google Scholar] [CrossRef]
  5. Huang, S.; Huang, G. Preparation and drug delivery of dextran-drug complex. Drug Deliv. 2019, 26, 252–261. [Google Scholar] [CrossRef] [Green Version]
  6. Zamani, R.; Bizari, D.; Heiat, M. Synthesis and characterization of phase shift dextran stabilized nanodroplets for ultrasound-induced cancer therapy: A novel nanobiotechnology approach. J. Biotechnol. 2022, 350, 17–23. [Google Scholar] [CrossRef]
  7. Mufamadi, M.S.; Pillay, V.; Choonara, Y.E.; Du Toit, L.C.; Modi, G.; Naidoo, D.; Ndesendo, V.M.K. A Review on Composite Liposomal Technologies for Specialized Drug Delivery. J. Drug Deliv. 2011, 2011, 939851. [Google Scholar] [CrossRef]
  8. Malekhosseini, S.; Rezaie, A.; Khaledian, S.; Abdoli, M.; Zangeneh, M.M.; Hosseini, A.; Behbood, L. Fabrication and characterization of hydrocortisone loaded Dextran-Poly Lactic-co-Glycolic acid micelle. Heliyon 2020, 6, e03975. [Google Scholar] [CrossRef]
  9. Shaki, H.; Ganji, F.; Kempen, P.J.; Dolatshahi-Pirouz, A.; Vasheghani-Farahani, E. Self-assembled amphiphilic-dextran nanomicelles for delivery of rapamycin. J. Drug Deliv. Sci. Technol. 2018, 44, 333–341. [Google Scholar] [CrossRef]
  10. Qu, J.; Liang, Y.; Shi, M.; Guo, B.; Gao, Y.; Yin, Z. Biocompatible conductive hydrogels based on dextran and aniline trimer as electro-responsive drug delivery system for localized drug release. Int. J. Biol. Macromol. 2019, 140, 255–264. [Google Scholar] [CrossRef]
  11. Rangel Euzcateguy, G.; Parajua-Sejil, C.; Marchal, P.; Chapron, D.; Averlant-Petit, M.C.; Stefan, L.; Pickaert, G.; Durand, A. Rheological investigation of the influence of dextran on the self-assembly of lysine derivatives in water/dimethylsulfoxide mixtures. Colloids Surf. A Physicochem. Eng. Asp. 2021, 625, 126908. [Google Scholar] [CrossRef]
  12. Zhou, L.L.; Zhou, L.L.; Wei, C.; Guo, R. A bioactive dextran-based hydrogel promote the healing of infected wounds via antibacterial and immunomodulatory. Carbohydr. Polym. 2022, 291, 119558. [Google Scholar] [CrossRef] [PubMed]
  13. Delvart, A.; Moreau, C.; D’Orlando, A.; Falourd, X.; Cathala, B. Dextran-based polyelectrolyte multilayers: Effect of charge density on film build-up and morphology. Colloids Surf. B Biointerfaces 2022, 210, 112258. [Google Scholar] [CrossRef] [PubMed]
  14. Lee, H.; Han, Y.; Park, J.H. Enhanced deposition of Fe(III)-tannic acid complex nanofilm by Fe(III)-embedded dextran nanocoating. Appl. Surf. Sci. 2022, 573, 151598. [Google Scholar] [CrossRef]
  15. Raganato, L.; Del Giudice, A.; Ceccucci, A.; Sciubba, F.; Casciardi, S.; Sennato, S.; Scipioni, A.; Masci, G. Self-assembling nanowires from a linear l,d-peptide conjugated to the dextran end group. Int. J. Biol. Macromol. 2022, 207, 656–665. [Google Scholar] [CrossRef]
  16. Joshy, K.S.; George, A.; Snigdha, S.; Joseph, B.; Kalarikkal, N.; Pothen, L.A.; Thomas, S. Novel core-shell dextran hybrid nanosystem for anti-viral drug delivery. Mater. Sci. Eng. C 2018, 93, 864–872. [Google Scholar] [CrossRef]
  17. Hyon, W.; Shibata, S.; Ozaki, E.; Fujimura, M.; Hyon, S.H.; Matsumura, K. Elucidating the degradation mechanism of a self-degradable dextran-based medical adhesive. Carbohydr. Polym. 2022, 278, 118949. [Google Scholar] [CrossRef]
  18. Moydeen, A.M.; Ali Padusha, M.S.; Aboelfetoh, E.F.; Al-Deyab, S.S.; El-Newehy, M.H. Fabrication of electrospun poly(vinyl alcohol)/dextran nanofibers via emulsion process as drug delivery system: Kinetics and in vitro release study. Int. J. Biol. Macromol. 2018, 116, 1250–1259. [Google Scholar] [CrossRef]
  19. Banerjee, S.; Szepes, M.; Dibbert, N.; Rios-Camacho, J.C.; Kirschning, A.; Gruh, I.; Dräger, G. Dextran-based scaffolds for in-situ hydrogelation: Use for next generation of bioartificial cardiac tissues. Carbohydr. Polym. 2021, 262, 117924. [Google Scholar] [CrossRef]
  20. Pawlak, A.; Michely, L.; Belbekhouche, S. Multilayer dextran derivative based capsules fighting bacteria resistant to Antibiotic: Case of Kanamycin-Resistant Escherichia coli. Int. J. Biol. Macromol. 2022, 200, 242–246. [Google Scholar] [CrossRef]
  21. Odeniyi, M.; Omoteso, O.; Adepoju, A.; Jaiyeoba, K. Starch nanoparticles in drug delivery: A review. Polym. Med. 2019, 48, 41–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Rodrigues, A.; Emeje, M. Recent applications of starch derivatives in nanodrug delivery. Carbohydr. Polym. 2012, 87, 987–994. [Google Scholar] [CrossRef]
  23. Zhong, H.; Gao, X.; Cheng, C.; Liu, C.; Wang, Q.; Han, X. The Structural Characteristics of Seaweed Polysaccharides and Their Application in Gel Drug Delivery Systems. Mar. Drugs 2020, 18, 658. [Google Scholar] [CrossRef]
  24. Oprea, M.; Voicu, S.I. Recent advances in composites based on cellulose derivatives for biomedical applications. Carbohydr. Polym. 2020, 247, 116683. [Google Scholar] [CrossRef] [PubMed]
  25. Huo, M.; Wang, H.; Zhang, Y.; Cai, H.; Zhang, P.; Li, L.; Zhou, J.; Yin, T. Co-delivery of silybin and paclitaxel by dextran-based nanoparticles for effective anti-tumor treatment through chemotherapy sensitization and microenvironment modulation. J. Control. Release 2020, 321, 198–210. [Google Scholar] [CrossRef] [PubMed]
  26. Yue, J.; He, L.; Tang, Y.; Yang, L.; Wu, B.; Ni, J. Facile design and development of photoluminescent graphene quantum dots grafted dextran/glycol-polymeric hydrogel for thermoresponsive triggered delivery of buprenorphine on pain management in tissue implantation. J. Photochem. Photobiol. B Biol. 2019, 197, 111530. [Google Scholar] [CrossRef]
  27. Yilmaz, M.T.; İspirli, H.; Taylan, O.; Taşdemir, V.; Sagdic, O.; Dertli, E. Characterisation and functional roles of a highly branched dextran produced by a bee pollen isolate Leuconostoc mesenteroides BI-20. Food Biosci. 2022, 45, 101330. [Google Scholar] [CrossRef]
  28. dos Santos Campos, F.; Cassimiro, D.L.; Crespi, M.S.; Almeida, A.E.; Daflon Gremião, M.P. Preparation and characterisation of Dextran-70 hydrogel for controlled release of praziquantel. Brazilian J. Pharm. Sci. 2013, 49, 75–83. [Google Scholar] [CrossRef] [Green Version]
  29. Jamwal, S.; Ram, B.; Ranote, S.; Dharela, R.; Chauhan, G.S. New glucose oxidase-immobilized stimuli-responsive dextran nanoparticles for insulin delivery. Int. J. Biol. Macromol. 2019, 123, 968–978. [Google Scholar] [CrossRef]
  30. Tiryaki, E.; Başaran Elalmış, Y.; Karakuzu İkizler, B.; Yücel, S. Novel organic/inorganic hybrid nanoparticles as enzyme-triggered drug delivery systems: Dextran and Dextran aldehyde coated silica aerogels. J. Drug Deliv. Sci. Technol. 2020, 56, 101517. [Google Scholar] [CrossRef]
  31. Han, J.; Hang, F.; Guo, B.; Liu, Z.; You, C.; Wu, Z. Dextran synthesized by Leuconostoc mesenteroides BD1710 in tomato juice supplemented with sucrose. Carbohydr. Polym. 2014, 112, 556–562. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, Y.; Maina, N.H.; Coda, R.; Katina, K. Challenges and opportunities for wheat alternative grains in breadmaking: Ex-situ- versus in-situ-produced dextran. Trends Food Sci. Technol. 2021, 113, 232–244. [Google Scholar] [CrossRef]
  33. Wang, J.; Zhao, X.; Tian, Z.; Yang, Y.; Yang, Z. Characterization of an exopolysaccharide produced by Lactobacillus plantarum YW11 isolated from Tibet Kefir. Carbohydr. Polym. 2015, 125, 16–25. [Google Scholar] [CrossRef] [PubMed]
  34. Maingret, V.; Chartier, C.; Six, J.L.; Schmitt, V.; Héroguez, V. Pickering emulsions stabilized by biodegradable dextran-based nanoparticles featuring enzyme responsiveness and co-encapsulation of actives. Carbohydr. Polym. 2022, 284, 119146. [Google Scholar] [CrossRef] [PubMed]
  35. Rosca, I.; Petrovici, A.R.; Peptanariu, D.; Nicolescu, A.; Dodi, G.; Avadanei, M.; Ivanov, I.C.; Bostanaru, A.C.; Mares, M.; Ciolacu, D. Biosynthesis of dextran by Weissella confusa and its In vitro functional characteristics. Int. J. Biol. Macromol. 2018, 107, 1765–1772. [Google Scholar] [CrossRef] [PubMed]
  36. Kasaai, M.R. Dilute solution properties and degree of chain branching for dextran. Carbohydr. Polym. 2012, 88, 373–381. [Google Scholar] [CrossRef]
  37. Padmanabhan, P.A.; Kim, D.S. Production of insoluble dextran using cell-bound dextransucrase of Leuconostoc mesenteroides NRRL B-523. Carbohydr. Res. 2002, 337, 1529–1533. [Google Scholar] [CrossRef]
  38. Paúrová, M.; Taboubi, O.; Šeděnková, I.; Hromádková, J.; Matouš, P.; Herynek, V.; Šefc, L.; Babič, M. Role of dextran in stabilization of polypyrrole nanoparticles for photoacoustic imaging. Eur. Polym. J. 2021, 157, 110634. [Google Scholar] [CrossRef]
  39. Hu, P.; Lei, Q.; Duan, S.; Fu, Y.; Pan, H.; Chang, C.; Zheng, Z.; Wu, Y.; Zhang, Z.; Li, R.; et al. In-situ formable dextran/chitosan-based hydrogels functionalized with collagen and EGF for diabetic wounds healing. Biomater. Adv. 2022, 136, 212773. [Google Scholar] [CrossRef]
  40. Bakrania, A.K.; Variya, B.C.; Madan, P.; Patel, S.S. Repeated dose 28-day oral toxicity study of DEAE-Dextran in mice: An advancement in safety chemotherapeutics. Regul. Toxicol. Pharmacol. 2017, 88, 262–272. [Google Scholar] [CrossRef]
  41. Zhang, M.; Huang, Y.; Pan, W.; Tong, X.; Zeng, Q.; Su, T.; Qi, X.; Shen, J. Polydopamine-incorporated dextran hydrogel drug carrier with tailorable structure for wound healing. Carbohydr. Polym. 2021, 253, 117213. [Google Scholar] [CrossRef] [PubMed]
  42. Chechushkov, A.; Zaitseva, N.; Vorontsova, E.; Kozhin, P.; Menshchikova, E.; Shkurupiy, V. Dextran loading protects macrophages from lipid peroxidation and induces a Keap1/Nrf2/ARE-dependent antioxidant response. Life Sci. 2016, 166, 100–107. [Google Scholar] [CrossRef] [PubMed]
  43. Nguyen, H.V.; Campbell, K.; Painter, G.F.; Young, S.L.; Walker, G.F. Data on the uptake of CpG-loaded amino-dextran nanoparticles by antigen-presenting cells. Data Br. 2021, 35, 106883. [Google Scholar] [CrossRef] [PubMed]
  44. Alibolandi, M.; Mohammadi, M.; Taghdisi, S.M.; Ramezani, M.; Abnous, K. Fabrication of aptamer decorated dextran coated nano-graphene oxide for targeted drug delivery. Carbohydr. Polym. 2017, 155, 218–229. [Google Scholar] [CrossRef]
  45. Park, G.S.; Hong, S.J.; Jung, B.K.; Lee, C.; Park, C.K.; Shin, J.H. The complete genome sequence of a lactic acid bacterium Leuconostoc mesenteroides ssp. dextranicum strain DSM 20484T. J. Biotechnol. 2016, 219, 3–4. [Google Scholar] [CrossRef]
  46. Du, R.; Pei, F.; Kang, J.; Zhang, W.; Wang, S.; Ping, W.; Ling, H.; Ge, J. Analysis of the structure and properties of dextran produced by Weissella confusa. Int. J. Biol. Macromol. 2022, 204, 677–684. [Google Scholar] [CrossRef]
  47. Robyt, J.F.; Kimble, B.K.; Walseth, T.F. The mechanism of dextransucrase action. Direction of dextran biosynthesis. Arch. Biochem. Biophys. 1974, 165, 634–640. [Google Scholar] [CrossRef]
  48. Robyt, J.F.; Yoon, S.H.; Mukerjea, R. Dextransucrase and the mechanism for dextran biosynthesis. Carbohydr. Res. 2008, 343, 3039–3048. [Google Scholar] [CrossRef]
  49. Lazić, M.L.; Veljković, V.B.; Vučetić, J.I.; Vrvić, M.M. Effect of pH and aeration on dextran production by Leuconostoc mesenteroides. Enzyme Microb. Technol. 1993, 15, 334–338. [Google Scholar] [CrossRef]
  50. Falconer, D.J.; Mukerjea, R.; Robyt, J.F. Biosynthesis of dextrans with different molecular weights by selecting the concentration of Leuconostoc mesenteroides B-512FMC dextransucrase, the sucrose concentration, and the temperature. Carbohydr. Res. 2011, 346, 280–284. [Google Scholar] [CrossRef]
  51. Schmid, J. Recent insights in microbial exopolysaccharide biosynthesis and engineering strategies. Curr. Opin. Biotechnol. 2018, 53, 130–136. [Google Scholar] [CrossRef]
  52. Pintado, A.I.E.; Ferreira, J.A.; Pintado, M.M.E.; Gomes, A.M.P.; Malcata, F.X.; Coimbra, M.A. Efficiency of purification methods on the recovery of exopolysaccharides from fermentation media. Carbohydr. Polym. 2020, 231, 115703. [Google Scholar] [CrossRef]
  53. Wang, Y.; Xie, C.; Pulkkinen, M.; Edelmann, M.; Chamlagain, B.; Coda, R.; Sandell, M.; Piironen, V.; Maina, N.H.; Katina, K. In situ production of vitamin B12 and dextran in soya flour and rice bran: A tool to improve flavour and texture of B12-fortified bread. LWT 2022, 161, 113407. [Google Scholar] [CrossRef]
  54. Petrovici, A.R.; Nicolescu, A.; Silion, M.; Roșca, I.; Ciolacu, D. Biopolymer biosynthesis by lactic acid bacteria strain in four different culture media. Rev. Roum. Chim. 2018, 63, 637–642. [Google Scholar]
  55. Dimofte, A.; Simionescu, N.; Petrovici, A.R.; Spiridon, I. Probiotic Properties of Weissella confusa PP29 on Hibiscus sabdariffa L. Media. Fermentation 2022, 8, 553. [Google Scholar] [CrossRef]
  56. Petrovici, A.R.; Roşca, I.; Dodi, G.; Nicolescu, A.; Avǎdanei, M.; Varganici, C.D.; Ciolacu, D. Effects of culture medium composition on biosynthesis of exopolysaccharides. Cellul. Chem. Technol. 2017, 51, 821–830. [Google Scholar]
  57. Zhu, Y.; Wang, C.; Jia, S.; Wang, B.; Zhou, K.; Chen, S.; Yang, Y.; Liu, S. Purification, characterization and antioxidant activity of the exopolysaccharide from Weissella cibaria SJ14 isolated from Sichuan paocai. Int. J. Biol. Macromol. 2018, 115, 820–828. [Google Scholar] [CrossRef]
  58. Du, R.; Qiao, X.; Zhao, F.; Song, Q.; Zhou, Q.; Wang, Y.; Pan, L.; Han, Y.; Zhou, Z. Purification, characterization and antioxidant activity of dextran produced by Leuconostoc pseudomesenteroides from homemade wine. Carbohydr. Polym. 2018, 198, 529–536. [Google Scholar] [CrossRef]
  59. Yilmaz, M.T.; İspirli, H.; Taylan, O.; Bilgrami, A.L.; Dertli, E. Structural and bioactive characteristics of a dextran produced by Lactobacillus kunkeei AK1. Int. J. Biol. Macromol. 2022, 200, 293–302. [Google Scholar] [CrossRef]
  60. Li, J.; Ai, L.; Xu, F.; Hu, X.; Yao, Y.; Wang, L. Structural characterization of exopolysaccharides from Weissella cibaria NC516.11 in distiller grains and its improvement in gluten-free dough. Int. J. Biol. Macromol. 2022, 199, 17–23. [Google Scholar] [CrossRef]
  61. Chen, H.; Wang, H.; Wei, Y.; Hu, M.; Dong, B.; Fang, H.; Chen, Q. Super-resolution imaging reveals the subcellular distribution of dextran at the nanoscale in living cells. Chin. Chem. Lett. 2022, 33, 1865–1869. [Google Scholar] [CrossRef]
  62. Konhäuser, M.; Kannaujiya, V.K.; Steiert, E.; Schwickert, K.; Schirmeister, T.; Wich, P.R. Co-encapsulation of l-asparaginase and etoposide in dextran nanoparticles for synergistic effect in chronic myeloid leukemia cells. Int. J. Pharm. 2022, 622, 121796. [Google Scholar] [CrossRef] [PubMed]
  63. Li, L.; Bai, Z.; Levkin, P.A. Boronate-dextran: An acid-responsive biodegradable polymer for drug delivery. Biomaterials 2013, 34, 8504–8510. [Google Scholar] [CrossRef] [PubMed]
  64. Bachelder, E.M.; Pino, E.N.; Ainslie, K.M. Acetalated Dextran: A Tunable and Acid-Labile Biopolymer with Facile Synthesis and a Range of Applications. Chem. Rev. 2017, 117, 1915–1926. [Google Scholar] [CrossRef]
  65. Kauffman, K.J.; Do, C.; Sharma, S.; Gallovic, M.D.; Bachelder, E.M.; Ainslie, K.M. Synthesis and characterization of acetalated dextran polymer and microparticles with ethanol as a degradation product. ACS Appl. Mater. Interfaces 2012, 4, 4149–4155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Meenach, S.A.; Kim, Y.J.; Kauffman, K.J.; Kanthamneni, N.; Bachelder, E.M.; Ainslie, K.M. Synthesis, optimization, and characterization of camptothecin-loaded acetalated dextran porous microparticles for pulmonary delivery. Mol. Pharm. 2012, 9, 290–298. [Google Scholar] [CrossRef]
  67. Wang, Z.; Gupta, S.K.; Meenach, S.A. Development and physicochemical characterization of acetalated dextran aerosol particle systems for deep lung delivery. Int. J. Pharm. 2017, 525, 264–274. [Google Scholar] [CrossRef] [Green Version]
  68. Chen, N.; Collier, M.A.; Gallovic, M.D.; Collins, G.C.; Sanchez, C.C.; Fernandes, E.Q.; Bachelder, E.M.; Ainslie, K.M. Degradation of acetalated dextran can be broadly tuned based on cyclic acetal coverage and molecular weight. Int. J. Pharm. 2016, 512, 147–157. [Google Scholar] [CrossRef] [Green Version]
  69. Borteh, H.M.; Gallovic, M.D.; Sharma, S.; Peine, K.J.; Miao, S.; Brackman, D.J.; Gregg, K.; Xu, Y.; Guo, X.; Guan, J.; et al. Electrospun acetalated dextran scaffolds for temporal release of therapeutics. Langmuir 2013, 29, 7957–7965. [Google Scholar] [CrossRef]
  70. Curcio, M.; Cirillo, G.; Paolì, A.; Naimo, G.D.; Mauro, L.; Amantea, D.; Leggio, A.; Nicoletta, F.P.; Iemma, F. Self-assembling Dextran prodrug for redox- and pH-responsive co-delivery of therapeutics in cancer cells. Colloids Surf. B Biointerfaces 2020, 185, 110537. [Google Scholar] [CrossRef]
  71. Jalalvandi, E.; Hanton, L.R.; Moratti, S.C. Preparation of a pH sensitive hydrogel based on dextran and polyhydrazide for release of 5-flurouracil, an anticancer drug. J. Drug Deliv. Sci. Technol. 2018, 44, 146–152. [Google Scholar] [CrossRef]
  72. Wu, S.; Yang, Y.; Wang, S.; Dong, C.; Zhang, X.; Zhang, R.; Yang, L. Dextran and peptide-based pH-sensitive hydrogel boosts healing process in multidrug-resistant bacteria-infected wounds. Carbohydr. Polym. 2022, 278, 118994. [Google Scholar] [CrossRef] [PubMed]
  73. Thambi, T.; You, D.G.; Han, H.S.; Deepagan, V.G.; Jeon, S.M.; Suh, Y.D.; Choi, K.Y.; Kim, K.; Kwon, I.C.; Yi, G.R.; et al. Bioreducible Carboxymethyl Dextran Nanoparticles for Tumor-Targeted Drug Delivery. Adv. Healthc. Mater. 2014, 3, 1829–1838. [Google Scholar] [CrossRef] [PubMed]
  74. Korcová, J.; Machová, E.; Filip, J.; Bystrický, S. Biophysical properties of carboxymethyl derivatives of mannan and dextran. Carbohydr. Polym. 2015, 134, 6–11. [Google Scholar] [CrossRef]
  75. Um, W.; Kumar, E.K.P.; Song, Y.; Lee, J.; An, J.Y.; Joo, H.; You, D.G.; Park, J.H. Carboxymethyl dextran-based nanocomposites for enhanced chemo-sonodynamic therapy of cancer. Carbohydr. Polym. 2021, 273, 118488. [Google Scholar] [CrossRef]
  76. Liu, G.; Hong, R.Y.; Guo, L.; Li, Y.G.; Li, H.Z. Preparation, characterization and MRI application of carboxymethyl dextran coated magnetic nanoparticles. Appl. Surf. Sci. 2011, 257, 6711–6717. [Google Scholar] [CrossRef]
  77. Das, M.; Oyarzabal, E.A.; Chen, L.; Lee, S.H.; Shah, N.; Gerlach, G.; Zhang, W.; Chao, T.H.H.; Van Den Berge, N.; Liu, C.; et al. One-pot synthesis of carboxymethyl-dextran coated iron oxide nanoparticles (CION) for preclinical fMRI and MRA applications. Neuroimage 2021, 238, 118213. [Google Scholar] [CrossRef]
  78. Li, J.; Zhou, Y.; Li, M.; Xia, N.; Huang, Q.; Do, H.; Liu, Y.N.; Zhou, F. Carboxymethylated dextran-coated magnetic iron oxide nanoparticles for regenerable bioseparation. J. Nanosci. Nanotechnol. 2011, 11, 10187–10192. [Google Scholar] [CrossRef]
  79. Gaowa, A.; Horibe, T.; Kohno, M.; Tabata, Y.; Harada, H.; Hiraoka, M.; Kawakami, K. Enhancement of anti-tumor activity of hybrid peptide in conjugation with carboxymethyl dextran via disulfide linkers. Eur. J. Pharm. Biopharm. 2015, 92, 228–236. [Google Scholar] [CrossRef]
  80. Vasić, K.; Knez, Ž.; Konstantinova, E.A.; Kokorin, A.I.; Gyergyek, S.; Leitgeb, M. Structural and magnetic characteristics of carboxymethyl dextran coated magnetic nanoparticles: From characterization to immobilization application. React. Funct. Polym. 2020, 148, 104481. [Google Scholar] [CrossRef]
  81. Cha, H.; Lee, S.; Hwan Kim, S.; Kim, H.; Lee, D.S.; Lee, H.S.; Lee, J.H.; Park, J.W. Increased susceptibility of IDH2-deficient mice to dextran sodium sulfate-induced colitis. Redox Biol. 2017, 13, 32–38. [Google Scholar] [CrossRef]
  82. Chen, Y.; Mohanraj, V.J.; Wang, F.; Benson, H.A.E. Designing chitosan-dextran sulfate nanoparticles using charge ratios. Aaps PharmSciTech 2007, 8, 131–139. [Google Scholar] [CrossRef]
  83. Zhao, Y.; Jiang, Y.; Wang, Q.; Sun, Y.; Huang, K.; Yao, Z. Rapid and sensitive detection of dextran sulfate sodium based on supramolecular self-assembly of a perylene diimide derivative in aqueous solution. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2022, 270, 120760. [Google Scholar] [CrossRef]
  84. Arafa, H.M.M.; Hemeida, R.A.; El-Bahrawy, A.I.M.; Hamada, F.M.A. Prophylactic role of curcumin in dextran sulfate sodium (DSS)-induced ulcerative colitis murine model. Food Chem. Toxicol. 2009, 47, 1311–1317. [Google Scholar] [CrossRef] [PubMed]
  85. Balaha, M.; Kandeel, S.; Elwan, W. Garlic oil inhibits dextran sodium sulfate-induced ulcerative colitis in rats. Life Sci. 2016, 146, 40–51. [Google Scholar] [CrossRef] [PubMed]
  86. Arigesavan, K.; Sudhandiran, G. Carvacrol exhibits anti-oxidant and anti-inflammatory effects against 1,2-dimethyl hydrazine plus dextran sodium sulfate induced inflammation associated carcinogenicity in the colon of Fischer 344 rats. Biochem. Biophys. Res. Commun. 2015, 461, 314–320. [Google Scholar] [CrossRef] [PubMed]
  87. Yao, J.; Wang, J.Y.; Liu, L.; Li, Y.X.; Xun, A.Y.; Zeng, W.S.; Jia, C.H.; Wei, X.X.; Feng, J.L.; Zhao, L.; et al. Anti-oxidant Effects of Resveratrol on Mice with DSS-induced Ulcerative Colitis. Arch. Med. Res. 2010, 41, 288–294. [Google Scholar] [CrossRef]
  88. Hong, C.O.; Rhee, C.H.; Pyo, M.C.; Lee, K.W. Anti-inflammatory effect of glucose-lysine Maillard reaction products on intestinal inflammation model in vivo. Int. Immunopharmacol. 2017, 52, 324–332. [Google Scholar] [CrossRef]
  89. Liu, D.; Gao, L.; Zhang, J.; Huo, X.; Ni, H.; Cao, L. Anti-inflammatory and Anti-oxidant Effects of Licorice Flavonoids on Ulcerative Colitis in Mouse Model. Chin. Herb. Med. 2017, 9, 358–368. [Google Scholar] [CrossRef]
  90. Seo, S.; Shin, J.S.; Lee, W.S.; Rhee, Y.K.; Cho, C.W.; Do Hong, H.; Lee, K.T. Anti-colitis effect of Lactobacillus sakei K040706 via suppression of inflammatory responses in the dextran sulfate sodium-induced colitis mice model. J. Funct. Foods 2017, 29, 256–268. [Google Scholar] [CrossRef]
  91. Asari, T.; Kikuchi, H.; Kawaguchi, S.; Sakuraba, H.; Yoshida, S.; Akemoto, Y.; Maeda, T.; Shinji, O.; Murai, Y.; Higuchi, N.; et al. Polygonum tinctorium leaves suppress sodium dextran sulfate-induced colitis through interleukin-10-related pathway. Biochem. Biophys. Rep. 2022, 30, 101272. [Google Scholar] [CrossRef] [PubMed]
  92. Wieber, A.; Selzer, T.; Kreuter, J. Characterisation and stability studies of a hydrophilic decapeptide in different adjuvant drug delivery systems: A comparative study of PLGA nanoparticles versus chitosan-dextran sulphate microparticles versus DOTAP-liposomes. Int. J. Pharm. 2011, 421, 151–159. [Google Scholar] [CrossRef]
  93. Wang, F.; Li, J.; Tang, X.; Huang, K.; Chen, L. Polyelectrolyte three layer nanoparticles of chitosan/dextran sulfate/chitosan for dual drug delivery. Colloids Surf. B Biointerfaces 2020, 190, 110925. [Google Scholar] [CrossRef] [PubMed]
  94. Lalani, J.; Misra, A. Gene Delivery Using Chemical Methods. In Challenges in Delivery of Therapeutic Genomics and Proteomics; Elsevier: Amsterdam, The Netherlands, 2011; pp. 127–206. ISBN 9780123849649. [Google Scholar]
  95. Siewert, C.; Haas, H.; Nawroth, T.; Ziller, A.; Nogueira, S.S.; Schroer, M.A.; Blanchet, C.E.; Svergun, D.I.; Radulescu, A.; Bates, F.; et al. Investigation of charge ratio variation in mRNA—DEAE-dextran polyplex delivery systems. Biomaterials 2019, 192, 612–620. [Google Scholar] [CrossRef]
  96. Ju, X.; Šmíd, B.; Johánek, V.; Khalakhan, I.; Yakovlev, Y.; Matolínová, I.; Matolín, V. Investigation of dextran adsorption on polycrystalline cerium oxide surfaces. Appl. Surf. Sci. 2021, 544, 148890. [Google Scholar] [CrossRef]
  97. Piazza, R.D.; da Nunes, E.S.; Viali, W.R.; da Silva, S.W.; Aragón, F.H.; Coaquira, J.A.H.; de Morais, P.C.; Marques, R.F.C.; Jafelicci, M. Magnetic nanohydrogel obtained by miniemulsion polymerization of poly(acrylic acid) grafted onto derivatized dextran. Carbohydr. Polym. 2017, 178, 378–385. [Google Scholar] [CrossRef]
  98. Curcio, M.; Diaz-Gomez, L.; Cirillo, G.; Concheiro, A.; Iemma, F.; Alvarez-Lorenzo, C. pH/redox dual-sensitive dextran nanogels for enhanced intracellular drug delivery. Eur. J. Pharm. Biopharm. 2017, 117, 324–332. [Google Scholar] [CrossRef]
  99. He, L.; Zheng, R.; Min, J.; Lu, F.; Wu, C.; Zhi, Y.; Shan, S.; Su, H. Preparation of magnetic microgels based on dextran for stimuli-responsive release of doxorubicin. J. Magn. Magn. Mater. 2021, 517, 167394. [Google Scholar] [CrossRef]
  100. Yu, K.; Yang, X.; He, L.; Zheng, R.; Min, J.; Su, H.; Shan, S.; Jia, Q. Facile preparation of pH/reduction dual-stimuli responsive dextran nanogel as environment-sensitive carrier of doxorubicin. Polymer 2020, 200, 122585. [Google Scholar] [CrossRef]
  101. Almeida, J.F.; Ferreira, P.; Alves, P.; Lopes, A.; Gil, M.H. Synthesis of a dextran based thermo-sensitive drug delivery system by gamma irradiation. Int. J. Biol. Macromol. 2013, 61, 150–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Pacelli, S.; Di Muzio, L.; Paolicelli, P.; Fortunati, V.; Petralito, S.; Trilli, J.; Casadei, M.A. Dextran-polyethylene glycol cryogels as spongy scaffolds for drug delivery. Int. J. Biol. Macromol. 2021, 166, 1292–1300. [Google Scholar] [CrossRef] [PubMed]
  103. Prusty, K.; Swain, S.K. Nano silver decorated polyacrylamide/dextran nanohydrogels hybrid composites for drug delivery applications. Mater. Sci. Eng. C 2018, 85, 130–141. [Google Scholar] [CrossRef] [PubMed]
  104. He, M.; Teng, F.; Chen, H.; Wu, C.; Huang, Y.Y.; Li, Y. Fabrication of soy protein isolate-succinic anhydride-dextran nanogels: Properties, performance, and controlled release of curcumin. LWT 2022, 160, 113259. [Google Scholar] [CrossRef]
  105. Zhang, Q.; Yue, W.; Zhao, D.; Chen, L.; Xu, Z.; Lin, D.; Qin, W. Preparation and characterization of soybean protein isolate-dextran conjugate-based nanogels. Food Chem. 2022, 384, 132556. [Google Scholar] [CrossRef] [PubMed]
  106. Matsushige, C.; Xu, X.; Miyagi, M.; Zuo, Y.Y.; Yamazaki, Y. RGD-modified dextran hydrogel promotes follicle growth in three-dimensional ovarian tissue culture in mice. Theriogenology 2022, 183, 120–131. [Google Scholar] [CrossRef] [PubMed]
  107. Zhao, Y.; Jalili, S. Dextran, as a biological macromolecule for the development of bioactive wound dressing materials: A review of recent progress and future perspectives. Int. J. Biol. Macromol. 2022, 207, 666–682. [Google Scholar] [CrossRef]
  108. Su, H.; Zheng, R.; Jiang, L.; Zeng, N.; Yu, K.; Zhi, Y.; Shan, S. Dextran hydrogels via disulfide-containing Schiff base formation: Synthesis, stimuli-sensitive degradation and release behaviors. Carbohydr. Polym. 2021, 265, 118085. [Google Scholar] [CrossRef]
  109. Dhaneshwar, S.S.; Kandpal, M.; Gairola, N.; Kadam, S.S. Dextran: A promising macromolecular drug carrier. Indian J. Pharm. Sci. 2006, 68, 705–714. [Google Scholar] [CrossRef] [Green Version]
  110. Garg, A.; Rai, G.; Lodhi, S.; Jain, A.P.; Yadav, A.K. In-vitro and in-vivo assessment of dextran-appended cellulose acetate phthalate nanoparticles for transdermal delivery of 5-fluorouracil. Drug Deliv. 2014, 23, 1525–1535. [Google Scholar] [CrossRef]
  111. Tiyaboonchai, W.; Woiszwillo, J.; Middaugh, C.R. Formulation and characterization of amphotericin B–polyethylenimine–dextran sulfate nanoparticles. J. Pharm. Sci. 2001, 90, 902–914. [Google Scholar] [CrossRef]
  112. Abid, M.; Naveed, M.; Azeem, I.; Faisal, A.; Faizan Nazar, M.; Yameen, B. Colon specific enzyme responsive oligoester crosslinked dextran nanoparticles for controlled release of 5-fluorouracil. Int. J. Pharm. 2020, 586, 119605. [Google Scholar] [CrossRef] [PubMed]
  113. Wu, L.; Zhang, L.; Shi, G.; Ni, C. Zwitterionic pH/redox nanoparticles based on dextran as drug carriers for enhancing tumor intercellular uptake of doxorubicin. Mater. Sci. Eng. C 2016, 61, 278–285. [Google Scholar] [CrossRef] [PubMed]
  114. Dai, F.; Du, M.; Liu, Y.; Liu, G.; Liu, Q.; Zhang, X. Folic acid-conjugated glucose and dextran coated iron oxide nanoparticles as MRI contrast agents for diagnosis and treatment response of rheumatoid arthritis. J. Mater. Chem. B 2014, 2, 2240–2247. [Google Scholar] [CrossRef] [PubMed]
  115. Kaewprapan, K.; Inprakhon, P.; Marie, E.; Durand, A. Enzymatically degradable nanoparticles of dextran esters as potential drug delivery systems. Carbohydr. Polym. 2012, 88, 875–881. [Google Scholar] [CrossRef]
  116. Ferrari, P.F.; Zattera, E.; Pastorino, L.; Perego, P.; Palombo, D. Dextran/poly-L-arginine multi-layered CaCO3-based nanosystem for vascular drug delivery. Int. J. Biol. Macromol. 2021, 177, 548–558. [Google Scholar] [CrossRef]
  117. Lungoci, A.L.; Pinteala, M.; Petrovici, A.R.; Rosca, I.; Turin-Moleavin, I.A.; Fifere, A. Biosynthesized dextran coated magnetic nanoparticles with antifungal activity. Rev. Roum. Chim. 2018, 63, 497–503. [Google Scholar]
  118. Anghelache, M.; Turtoi, M.; Petrovici, A.R.; Fifere, A.; Pinteala, M.; Calin, M. Development of Dextran-Coated Magnetic Nanoparticles Loaded with Protocatechuic Acid for Vascular Inflammation Therapy. Pharmaceutics 2021, 13, 1414. [Google Scholar] [CrossRef]
  119. Wasiak, I.; Kulikowska, A.; Janczewska, M.; Michalak, M.; Cymerman, I.A.; Nagalski, A.; Kallinger, P.; Szymanski, W.W.; Ciach, T. Dextran nanoparticle synthesis and properties. PLoS ONE 2016, 11, e0146237. [Google Scholar] [CrossRef] [Green Version]
  120. Zhang, M.; Liu, J.; Kuang, Y.; Li, Q.; Zheng, D.W.; Song, Q.; Chen, H.; Chen, X.; Xu, Y.; Li, C.; et al. Ingenious pH-sensitive dextran/mesoporous silica nanoparticles based drug delivery systems for controlled intracellular drug release. Int. J. Biol. Macromol. 2017, 98, 691–700. [Google Scholar] [CrossRef]
  121. Zhu, J.Y.; Lei, Q.; Yang, B.; Jia, H.Z.; Qiu, W.X.; Wang, X.; Zeng, X.; Zhuo, R.X.; Feng, J.; Zhang, X.Z. Efficient nuclear drug translocation and improved drug efficacy mediated by acidity-responsive boronate-linked dextran/cholesterol nanoassembly. Biomaterials 2015, 52, 281–290. [Google Scholar] [CrossRef]
  122. Wu, F.; Zhou, Z.; Su, J.; Wei, L.; Yuan, W.; Jin, T. Development of dextran nanoparticles for stabilizing delicate proteins. Nanoscale Res. Lett. 2013, 8, 197. [Google Scholar] [CrossRef] [Green Version]
  123. Kesharwani, P.; Gorain, B.; Low, S.Y.; Tan, S.A.; Ling, E.C.S.; Lim, Y.K.; Chin, C.M.; Lee, P.Y.; Lee, C.M.; Ooi, C.H.; et al. Nanotechnology based approaches for anti-diabetic drugs delivery. Diabetes Res. Clin. Pract. 2018, 136, 52–77. [Google Scholar] [CrossRef] [PubMed]
  124. El Founi, M.; Soliman, S.M.A.; Vanderesse, R.; Acherar, S.; Guedon, E.; Chevalot, I.; Babin, J.; Six, J.L. Light-sensitive dextran-covered PNBA nanoparticles as triggered drug delivery systems: Formulation, characteristics and cytotoxicity. J. Colloid Interface Sci. 2018, 514, 289–298. [Google Scholar] [CrossRef] [PubMed]
  125. Medhat, D.; Hussein, J.; El-Naggar, M.E.; Attia, M.F.; Anwar, M.; Latif, Y.A.; Booles, H.F.; Morsy, S.; Farrag, A.R.; Khalil, W.K.B.; et al. Effect of Au-dextran NPs as anti-tumor agent against EAC and solid tumor in mice by biochemical evaluations and histopathological investigations. Biomed. Pharmacother. 2017, 91, 1006–1016. [Google Scholar] [CrossRef] [PubMed]
  126. El-Sayed, N.S.; Sajid, M.I.; Parang, K.; Tiwari, R.K. Synthesis, characterization, and cytotoxicity evaluation of dextran-myristoyl-ECGKRK peptide conjugate. Int. J. Biol. Macromol. 2021, 191, 1204–1211. [Google Scholar] [CrossRef]
  127. Kashyap, A.; Kaur, R.; Baldi, A.; Jain, U.K.; Chandra, R.; Madan, J. Chloroquine diphosphate bearing dextran nanoparticles augmented drug delivery and overwhelmed drug resistance in Plasmodium falciparum parasites. Int. J. Biol. Macromol. 2018, 114, 161–168. [Google Scholar] [CrossRef]
  128. Sampath, M.; Pichaimani, A.; Kumpati, P.; Sengottuvelan, B. The remarkable role of emulsifier and chitosan, dextran and PEG as capping agents in the enhanced delivery of curcumin by nanoparticles in breast cancer cells. Int. J. Biol. Macromol. 2020, 162, 748–761. [Google Scholar] [CrossRef]
  129. Remya, N.S.; Syama, S.; Sabareeswaran, A.; Mohanan, P.V. Toxicity, toxicokinetics and biodistribution of dextran stabilized Iron oxide Nanoparticles for biomedical applications. Int. J. Pharm. 2016, 511, 586–598. [Google Scholar] [CrossRef]
  130. Bai, S.; Gao, Y.E.; Ma, X.; Shi, X.; Hou, M.; Xue, P.; Kang, Y.; Xu, Z. Reduction stimuli-responsive unimolecular polymeric prodrug based on amphiphilic dextran-framework for antitumor drug delivery. Carbohydr. Polym. 2018, 182, 235–244. [Google Scholar] [CrossRef]
  131. Chen, Z.; Krishnamachary, B.; Bhujwalla, Z.M. Degradable dextran nanopolymer as a carrier for choline kinase (Chok) siRNA cancer therapy. Nanomaterials 2016, 6, 34. [Google Scholar] [CrossRef] [Green Version]
  132. Bhaw-Luximon, A.; Goonoo, N.; Jhurry, D. Nanotherapeutics promises for colorectal cancer and pancreatic ductal adenocarcinoma. In Nanobiomaterials in Cancer Therapy: Applications of Nanobiomaterials; William Andrew Publishing: Norwich, NY, USA, 2016; pp. 147–201. ISBN 9780323428866. [Google Scholar]
  133. Manju, S.; Sreenivasan, K. Functionalised nanoparticles for targeted drug delivery. In Biointegration of Medical Implant Materials: Science and Design; Woodhead Publishing: Sawston, UK, 2010; pp. 267–297. ISBN 9781845695095. [Google Scholar]
  134. Jiang, L.; Li, S.; Wang, N.; Zhao, S.; Chen, Y.; Chen, Y. Preparation of dextran-casein phosphopeptide conjugates, evaluation of its calcium binding capacity and digestion in vitro. Food Chem. 2021, 352, 129332. [Google Scholar] [CrossRef] [PubMed]
  135. Parhizkar, E.; Samani, S.M.; Sakhteman, A.; Daneshamouz, S.; Parhizkar, G.; Ahmadi, F. Synthesis, cytotoxicity assay, pharmacokinetics, biodistribution and modeling study of cabazitaxel-dextran nanoconjugates: Targeted vs non targeted delivery. Colloids Surf. B Biointerfaces 2022, 209, 112187. [Google Scholar] [CrossRef] [PubMed]
  136. Kim, W.; Yang, Y.; Kim, D.; Jeong, S.; Yoo, J.W.; Yoon, J.H.; Jung, Y. Conjugation of metronidazole with dextran: A potential pharmaceutical strategy to control colonic distribution of the anti-amebic drug susceptible to metabolism by colonic microbes. Drug Des. Devel. Ther. 2017, 11, 419–429. [Google Scholar] [CrossRef] [PubMed]
  137. Zhang, X.; Zhang, T.; Ma, X.; Wang, Y.; Lu, Y.; Jia, D.; Huang, X.; Chen, J.; Xu, Z.; Wen, F. The design and synthesis of dextran-doxorubicin prodrug-based pH-sensitive drug delivery system for improving chemotherapy efficacy. Asian J. Pharm. Sci. 2020, 15, 605–616. [Google Scholar] [CrossRef] [PubMed]
  138. Jin, R.; Guo, X.; Dong, L.; Xie, E.; Cao, A. Amphipathic dextran-doxorubicin prodrug micelles for solid tumor therapy. Colloids Surf. B Biointerfaces 2017, 158, 47–56. [Google Scholar] [CrossRef]
  139. Zhang, B.; Gao, Y.; Zhang, X.; Jiang, J.; Ren, J.; Wang, S.; Hu, H.; Zhao, Y.; Chen, L.; Zhao, K.; et al. Ultra-stable dextran conjugated prodrug micelles for oxidative stress and glycometabolic abnormality combination treatment of Alzheimer’s disease. Int. J. Biol. Macromol. 2022, 203, 430–444. [Google Scholar] [CrossRef]
Table 1. Biosynthesized DEX amount and molecular mass depending on culture media composition.
Table 1. Biosynthesized DEX amount and molecular mass depending on culture media composition.
StrainCulture MediaFermentation ConditionsDry DEX Amount, g/LMolecular Mass, DaReferences
Leuconostoc mesenteroides ZDRAVLJE
SR-P
Sucrose, yeast extract, barley malt extract, Na2HPO4 • 12 H2O, MgSO4 • 7 H2O, KCI, supplemented with 12% sucrose200 rpm54.9 [49]
Leuconostoc mesenteroides BD1710Tomato juice with 15% sucrose48 h at 28 °C32.06.35 × 105[31]
Weissella confusa PP29MRS, sucrose (80) dissolved in UHT milk48 h at 33 °C25.2 1.2 × 106[35]
LAB-PP15MRS, sucrose (80) dissolved in UHT milk48 h at 33 °C, 100 rpm9.01.9 × 105[56]
W. confusa H2MRS48 h at 30 °C 2.705 × 106[46]
W. cibaria
SJ14
Modified MRS semi-defined medium34 h at 37 °C0.337.12 × 104[57]
Leu. pseudomesenteroides
DRP-5
MRS agar36 h at 30 °C 6.23 × 106[58]
Leuconostoc mesenteroides BI-20, FYP broth with 3% sucrose48 h at 30 °C 1 × 108[27]
Weissella confusa A16Soya flour or rice bran with 10% sucrose24 h at 25 °C, 150 rpm58.0 [53]
Lactobacillus kunkeei AK1FYP broth with 3% sucrose48 h at 30 °C 45 × 103[59]
Weissella cibaria NC516.11Distiller grains of Fenjiu24–48 h at 37 °C 2.82 × 106[60]
Table 2. Dextran applications in drug-delivery systems.
Table 2. Dextran applications in drug-delivery systems.
DDS TypeDrug LoadedTargeted Disease/ApplicationsObservationsReference
HydrogelPolydopamineMultidrug-resistant bacterial infectionsGood physical and chemical properties; low cytotoxicity against mouse fibroblast cells; precise in vivo antibacterial and wound-healing performance[41]
Nanohydrogel matrix Maghemite Magnetic properties; high drug loading and stability in the circulatory system[97]
HydrogelAniline trimer elastomerSmart DDS for localised drug releaseControllable swelling ratio; stable rheological properties; good conductivity; electric stimuli-dependent activity[10]
NanogelMethotrexateHeLa cellsSensitive to the variation of the pH and redox environment; high release rate at pH 5.0; suitable carriers for cancer chemotherapeutics[98]
Magnetic microgelsDoxorubicin Promising results for further studies[99]
NanogelsDoxorubicinH1299 cancer cell lineThe indisputable results promote this system for further in vivo testing[100]
HydrogelsPraziquantelAnthelmintic disease Good in vitro results[28]
HydrogelsOndansetronTMAntiemetic following chemotherapyGood release kinetics’ curve[101]
CryogelsVitamin B12Vitamin B12 deficienciesSuitable carriers for water-soluble biomolecules’ delivery[102]
Micro-hydrogelIndole;
3-nitrophenol;
hydroxybenzoic acid;
diclofenac;
Very satisfactory release kinetics’ curve[2]
NanohydrogelsOrnidazoleClostridium sp. infectionsVery good in vitro antibiotic effect[103]
NanogelCurcuminNew foods developmentIn vitro simulations showed sustained drug release[104]
Nanogel Food ingredient preparationHigh potential for hydrophobic bioactive compounds’ encapsulation[105]
HydrogelsArginine-glycine-aspartic acid (RGD) sequencesArtificial cardiac tissuesPromising system for building cardiac grafts[19]
HydrogelsRGD and activin AOvarian tissue cultureSignificantly improves follicular oocytes’ in vitro maturation and development; synergistic effects in 3D tissue culture development[106]
Table 3. DEX-based NPs developed for drug delivery in different pathologies.
Table 3. DEX-based NPs developed for drug delivery in different pathologies.
DDS TypeDrug LoadedTargeted Disease/ApplicationObservationsReference
NPLidocaine Very good drug-release results[115]
NPModel protein and antibodiesCardiovascular pathologiesA promising tool for further in vivo tests[116]
Magnetic NP-DEX coatedProtocatechuic acidVascular inflammationVery good in silico results[117]
Magnetic NP-DEX coatedProtocatechuic acidVascular inflammationVery good in vitro results[118]
NP5-fluorouracilSkin damageLess immunogenic compared with other systems[110]
NP5-fluorouracilColorectal cancerThe HCT116 colon cancer cell line treatment was efficient.[112]
NPDoxorubicin pH/redox-responsive, self-assembly in aqueous solutions;
excellent plasmatic stability and anti-protein adsorption ability for tumour cellular uptake.
[113]
NPDodecilamine and doxorubicin pH-sensitive drug release[119]
NPDoxorubicin pH-sensitive intracellular drug release in HeLa cells[120]
NPDoxorubicin Acid-responsive NP in water;
loaded system toxicity on HeLa cells is comparable to the drug’s;
[63]
NPDoxorubicinHuman cervix carcinoma cells (HeLa)No DDS cytotoxicity and structural stability under the simulated physiological conditions;
drug release in acidic conditions;
very good in vivo results
[121]
NPAmphotericin BCandida albicans infectionNo loaded DDS toxicity compared with free drug.
Very good results
[111]
NPBovine serum albumin, granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor (G-CSF), β-galactosidase and myoglobinProtein stabilization for pharmaceuticals applicationsDEX NPs can preserve the protein’s bioactivity during the preparation process;
DEX NPs attenuate the acidic microenvironment by means of the dilution effect;
[122]
NPInsulinDiabetesVery good results[123]
Magnetic NP-DEX coatedPropiconazole Candida albicans infectionDirect interaction with the cell wall in both planktonic and biofilm phases;
77% biofilm breakdown
[117]
Magnetic NP-DEX coatedFolic acidMagnetic resonance imagingNegative contrast agent for antigen allowed arthritis visualisation in a rat model and measuring the treatment response[114]
NP-DEX coated Human epithelial colorectal adenocarcinoma cellsGood anticancer effect[124]
Gold NP-DEX coated Solid carcinoma and Ehrlich ascites carcinoma transplanted on miceSignificant antitumour effects;
Improvement of body functions;
increased liver antioxidant properties;
increased the B-cell lymphoma 2 gene expression level;
suppressed the apoptotic pathway
[125]
NP-DEX coatedZidovudineViral infectionIncreased drug half-life;
well internalized in the neural cells
[16]
NPMyristoyl-
ECGKRK peptide
Cancer therapiesSatisfactory results obtained[126]
NPChloroquine diphosphatePlasmodium falciparum malaria infectionVery good antimicrobial effects obtained
DDS suitable for in vivo tests
[127]
NPCurcuminBreast cancer DDS has good drug-loading and delivery performance; very effective against MCF-7 cell line[128]
Table 4. DEX-based NCs developed for drug delivery.
Table 4. DEX-based NCs developed for drug delivery.
DDS TypeDrug LoadedTargeted Disease/ApplicationsObservationsReference
NCCamptothecin Cancer therapiesHigh drug-loading rate; superior stability in aqueous solutions;
notable in vitro antitumour activity against HeLa and MCF-7 cells
[130]
NCCholine kinase siRNAsiRNA cancer therapy Successful delivery of siRNA[131]
DEX-coated graphene oxide NPCurcuminMCF-7 breast cancer cell linesVery good results obtained; potential DDS for chemotherapy application[44]
NCPaclitaxel and silybinA549 lung cancer cellsExcellent encapsulation efficiency of both active substances;
employs synergistic effects through chemotherapy sensitization and microenvironment modulation, improving the efficacy of cancer therapy; in vivo tests confirmed tumour growth inhibition
[25]
ConjugateCalcium ionsCalcium supplements’ carrier Could be used as an effective carrier for new calcium supplements[134]
Nanowires Pharmaceutical applicationsUseful biomaterial for medical applications[15]
NCCabazitaxelProstatic cancerPromising DDS as a substitution for the current market formulation[135]
ConjugateMetronidazole Protozoa infectionVery good in vivo results[136]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Petrovici, A.R.; Pinteala, M.; Simionescu, N. Dextran Formulations as Effective Delivery Systems of Therapeutic Agents. Molecules 2023, 28, 1086. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules28031086

AMA Style

Petrovici AR, Pinteala M, Simionescu N. Dextran Formulations as Effective Delivery Systems of Therapeutic Agents. Molecules. 2023; 28(3):1086. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules28031086

Chicago/Turabian Style

Petrovici, Anca Roxana, Mariana Pinteala, and Natalia Simionescu. 2023. "Dextran Formulations as Effective Delivery Systems of Therapeutic Agents" Molecules 28, no. 3: 1086. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules28031086

Article Metrics

Back to TopTop