Next Article in Journal
Effects of Overlap Length on Flammability and Fire Hazard of Vertical Polymethyl Methacrylate (PMMA) Plate Array
Next Article in Special Issue
Engineering of Doxorubicin-Encapsulating and TRAIL-Conjugated Poly(RGD) Proteinoid Nanocapsules for Drug Delivery Applications
Previous Article in Journal
Bicomponent Carbon Fibre within Woven Fabric for Protective Clothing
Previous Article in Special Issue
Tailoring Midazolam-Loaded Chitosan Nanoparticulate Formulation for Enhanced Brain Delivery via Intranasal Route
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nanocellulose in Drug Delivery and Antimicrobially Active Materials

1
Laboratory for Separation Processes and Product Design, Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova ulica 17, SI-2000 Maribor, Slovenia
2
Faculty of Mechanical Engineering, University of Maribor, Smetanova ulica 17, SI-2000 Maribor, Slovenia
3
Faculty of Medicine, University of Maribor, Taborska ulica 8, SI-2000 Maribor, Slovenia
*
Author to whom correspondence should be addressed.
Polymers 2020, 12(12), 2825; https://doi.org/10.3390/polym12122825
Submission received: 26 October 2020 / Revised: 25 November 2020 / Accepted: 26 November 2020 / Published: 27 November 2020
(This article belongs to the Special Issue Polymers and Drug Delivery Systems)

Abstract

:
In recent years, nanocellulose (NC) has also attracted a great deal of attention in drug delivery systems due to its unique physical properties, specific surface area, low risk of cytotoxicity, and excellent biological properties. This review is focused on nanocellulose based systems acting as carriers to be used in drug or antimicrobial delivery by providing different but controlled and sustained release of drugs or antimicrobial agents, respectively, thus showing potential for different routes of applications and administration. Microorganisms are increasingly resistant to antibiotics, and because, generally, the used metal or metal oxide nanoparticles at some concentration have toxic effects, more research has focused on finding biocompatible antimicrobial agents that have been obtained from natural sources. Our review contains the latest research from the last five years that tested nanocellulose-based materials in the field of drug delivery and antimicrobial activity.

Graphical Abstract

1. Introduction

Cellulose is a major component of several plant tissues and, as such, the most abundant renewable polymer resource available [1,2]. It is a linear polymer of D-anhydroglucopyranose units (AGUs), connected by β-(1->4)-glycosidic bonds, where a repeat unit is a dimer of glucose, cellobiose (Figure 1) [3,4].
Nanocellulose, in the form of fibrils (CNF) or crystals (CNC) [5,6], can be obtained from four main natural sources (Figure 2): bacteria, plants, algae, and animals [7].
While CNCs are prepared via acid hydrolysis [15,16,17], CNFs can be obtained by hydrolysis, oxidation and/or mechanical decomposition of biomass [18,19,20], or are produced by Gram-negative bacteria (mainly from Gluconacetobacter xylinus [21,22,23,24]) (Bacterial nanocellulose; BNC) [25], forming a highly viscous hydrogel when being suspended in water [26,27]. Among them, bacterial cellulose (BC) is the purest, contains no byproducts or contaminant molecules such as lignin, pectin, and hemicelluloses [28,29]. Its mechanical properties, ultrafine and highly porous network structure with high (over 95%) water-holding capacity, large surface area, and excellent biocompatibility [30] are its main advantages as compared to the plant-extracted NC, showing its extensive usage in the Cosmetic industry [31].
High strength and stiffness combined with low weight [32] and low risk of cytotoxicity [6,33] (Figure 3), as well as the ability to be formed into larger two-dimensional (2D) and three-dimensional (3D) structures, including membranes, films, porous aerogels and foams [34,35,36,37,38], are other outstanding properties of NC. Its application has thus received increasing attention in many applications, besides the paper industry also in (bio) medicinal, pharmaceutical, cosmetics and the food industry [39,40,41,42,43]. Furthermore, the use and addition of nanocellulose as a drug carrier could, simultaneously, target the local drug delivery to reduce consumption and control the release of the incorporated drug [44]. Because of NCs high surface to volume ratio, a higher cellular binding and uptake is provided, which causes an increase in the effectiveness of such delivery systems [45]. The application of both external and internal delivery systems of nanocellulose-based drugs is possible [46].
An abundance of infectious diseases is occurring, and besides, antibiotic resistance is increasing. Therefore, it is necessary to search for new effective surface disinfection and alternative materials that strive for antimicrobial and other bioactive characteristics. The traumatic injuries and burn wound healing process can be extended if bacteria cause wound infection. Hence, particularly interesting are products derived from natural sources that would have antimicrobial properties and would inhibit the growth and reproduction of microorganisms [6,47].
It is known that most of the bacterial cell walls are negatively charged. Indeed, some compounds (quaternary ammonium compounds, molecules, or polymers) could interact electrostatically with the negatively charged bacterial cell wall. Therefore, it causes membrane disruption and subsequent posterior death [48,49]. Tissues based on cellulose are often impregnated with active substances (e.g., essential oils, plant extracts, etc.), which are bound inside the cellulose network. The abundance of hydroxyl groups in such a structure is perfect for active substances and water molecules to bond inside the hydrogen-bonded network, thus allowing these substances to bind and then transfer into the skin [50]. The hydroxyl groups on the surface and the relatively large specific surface area of NC also provide many options for its modification and functionalization. To obtain functional groups to the nanocellulose surface, and to use these compounds as precursors for further applications, covalent modifications (e.g., oxidation, esterification, etherification, polymerization, etc.) are usually used, as well as non-covalent binding of the functional molecules [51,52,53,54].
Natural polymers such as starch, gelatin, chitosan, collagen, and cellulose, have enormous proven potential in the Biomedicine field in recent years, with many studies [6,55]. In Biomedicine, biocompatibility with no toxic effect, immunological rejection and/or physiological reaction [56,57,58,59] is one of the most important features [60,61,62,63]. For healing of wound infections, it is beneficial if the material has a porous network structure because this enables the potential transfer of antibiotics or other medicines into the wound, as well as helps to prevent additional infection, and serves as a physical barrier for other microorganisms [47]. NC has an ideal structure but has no antimicrobial activity itself. Therefore, it has been combined with other biologically active molecules, including other polysaccharides, proteins, glycosides, cytokines, growth factors, local anesthetics, and even nanoparticles [26,33,64].
Depending on the used antimicrobial agent for conjunction, the nanocellulose-based antimicrobial materials are, thus, divided into two groups [65], depending on the type of incorporated antimicrobial agents, i.e., inorganic [23,66,67,68,69,70] and organic [71,72,73,74,75,76,77] materials.
This article is, thus, a review of studies published over the last five years on nanocellulose-based drug and antimicrobial agent delivery systems, by covering both “standard” inorganic drugs and antimicrobial agents (e.g., silver nanoparticles), as well as alternatives to them that are obtained from natural sources and exhibit better biocompatibility and lower toxicity. Such nanocellulose based natural hybrids also have a high potential to reduce microbial growth and, thus, could be further used in food, medical and pharmaceutical applications, as well as may help to prevent the spread of various infectious diseases.

2. Application of Nanocellulose in Drug Delivery

A drug can be delivered by swallowing, by inhalation, by absorption, through the skin, or by intravenous injection to produce a systemic pharmacological effect. The oral route is the most common route of drug delivery to patients, where a drug is swallowed. The problem, however, is that it is covered by low bioavailability and poor absorption of protein and peptide drugs, for what is responsible for enzymatic degradation and high molecular composition of these drugs, which results in low membrane permeability [78]. Drugs are usually retained for a short period on the mucous membranes, and the rate of diffusion through the mucous surfaces is limited, so the bioavailability of the drugs is lower. One of the key factors is the amount of time the active substance must be absorbed. Mucoadhesive drug delivery systems are often used to improve the therapeutic efficacy of the drugs. For an active substance to be absorbed in the body, it must pass through mucous membranes. Well-known routes of mucoadhesive drug delivery systems are buccal, nasal, ocular, gastro, vaginal, and rectal [79,80,81]. Further, inhalation therapy is one of the best options for pulmonary delivery as a route for systemic administration. Aerosols are used for the delivery of therapeutic agents [82]. Intravenous infusion is also commonly used for the delivery of analgesics, sedatives, antibiotics, chemotherapy agents, hormones, anesthetics, and other fluids to patients [83]. Delivery of drugs via the skin, the transdermal route, has many advantages, such as a relatively large area for absorption, less frequent dosing, noninvasive nature and avoidance of first-pass metabolism [84]. The outer layer of the skin acts as a physical barrier, so only particular molecules with suitable physicochemical properties can penetrate the skin [85]. The solution to increasing the rate of drug delivery across the skin is chemical penetration enhancers, which are also often involved in improving the bioavailability of drugs [86,87].
It is well known that cellulose has been used successfully in the US Food and Drug Administration (FDA) approved drugs or products [88]. Due to the essential properties of nanocellulose, such as high crystallinity, biocompatibility, biodegradability, high surface area, unique mechanical and rheological properties, morphology and geometrical dimensions [2,17,89,90,91], it has become widely researched for drug delivery systems in recent years. The potential multifunctionality as regards chemical modifications could be used to bind and release various therapeutic agents [92,93]. Over the last decade, nanocellulose has become extremely attractive and used in many drug delivery studies. CNFs, CNCs, and BC are quite similar, but based on certain differences and characteristics, each type of NC is better suited for a particular drug delivery system [94]. Moreover, the release time of NC-based systems varies from a few minutes to several days or months [6,95,96].
In 2014, Jorfi and Foster [55] published a review of advances in NC for biomedical applications and have covered many important applications of NC in drug delivery systems by then. Further, Plackett et al. [97] published a review of NC as a vehicle for drug delivery by covering CNC, CNF, and BC in drug delivery systems published until 2014. Later in 2019, Kiliona et al. [98] collected various modification agents for CNC and CNF-based delivery systems with different model drugs, entrapping interactions and potential applications. In addition, Salimi et al. [96] gathered literature about NC applications in oral, transdermal and local drug delivery. Because things are moving very fast in this research area, Section 2.1, Section 2.2, and Section 2.3 summarize the applications (Table 1, Table 2 and Table 3) of NC in drug delivery systems over the last five years.

2.1. CNCs as a Vehicle for Drug Delivery

Lin et al. [99] developed biocompatible double-membrane hydrogels made from CNCs and alginate, according to the preparation method depicted in Figure 4.
Such hydrogels can release one drug quickly and another drug slowly, or, in other words, co-delivery of drugs is possible. For potential new anticancer drug delivery systems, Ntoutoume et al. [100] developed complexes from CNCs and cyclodextrin and loaded them with curcumin. Developed complexes have an antiproliferative effect on colon and rectal cancer cell lines. You et al. [101] cross-linked nanocomposite hydrogels based on quaternized cellulose (QC) and cationic CNCs with β-glycerophosphate (β-GP). Doxorubicin has been encapsulated into QC/CNC/β-GP hydrogels, and such hydrogels exhibited potential for applications in the subcutaneous delivery of anticancer drugs. Golshan et al. [102] prepared poly(propylene imine) dendrimer-grafted CNC nanostructures and conjugated these NPs with folic acid (FA) to investigate its bioconjugation effect on doxorubicin release behavior. FA conjugation prevented doxorubicin molecules from leaving the nanoconjugate system. Gorgieva et al. [103] conjugated sodium alendronate (Aln) and 3-aminopropylphosphoric acid (ApA) covalently to CNCs. These molecules contain bisphosphonates groups, which are used in drugs for the treatment of some bone diseases. With confocal microscopy, fluorescent labeling was performed with Rhodamine B Iso Thiocyanate (RBITC). Aln/Apa-modified CNCs did show much potential in drug delivery for bone cell-related diseases. Supramaniam et al. [104] synthesized magnetic CNCs (m-CNCs) and merged them with alginate. The hydrogel was loaded with ibuprofen as a model drug. The physical and mechanical properties of alginate hydrogel beads improved due to the presence of m-CNCs, which was reflected in the increased swelling degree and decreased ibuprofen release rate. In another study [105], poly(vinyl alcohol) (PVA) was reinforced with CNCs to obtain self-standing hydrogels. To prove the possibility of using hydrogels in applications for ophthalmic use, hydrogel lenses were loaded with chitosan-poly(acrylic acid) NPs and exposed to lysozyme, which is present in the eye. Md Abu et al. [106] studied the use of NC as an antimicrobial drug delivery system for honey and its application for wound healing. Due to released kinetics and good antimicrobial activity, honey incorporated CNC films could be used as wound dressings.
Many studies did investigate the binding of curcumin on NC materials. For example, Zainuddin et al. [107] extracted CNCs from the kenaf bast fibers and modified them with the cationic surfactant cetyltrimethylammonium bromide (CTAB). Further, curcumin was bound so that the curcumin/CTAB-CNC suspension was centrifuged, and a pellet was formed. It was found that the amount of curcumin bound was decreased with increasing the concentration of CTAB. Tong and colleagues [108] studied the use of CNC films for antimicrobial drug delivery in a diabetic wound dressing. Curcumin was added for the long-lasting antimicrobial effect of the film. Further, Gunathulake, Ching and colleagues [109,110,111] loaded curcumin into chitosan/CNC hydrogel. The nonionic surfactant medium (Tween 20) increased the drug loading capacity of NC compared to the drug loading capacity in the methanolic medium. For preserving drug activity, it is crucial that curcumin retains its structural integrity after release to simulated gastric fluid (SGF) and phosphate-buffered saline (PBS).
Furthermore, Li et al. [112] prepared CNCs with folate, cis-acetonitrile-doxorubicin, and polyethyleneimine. Hybrids released 95% of doxorubicin in 24 h at pH 5.5. Hivechi et al. [113] synthesized CNC-incorporated poly(ε-caprolactone) (PCL) nanofibers and studied its drug release behavior. Tetracycline was used as a model drug, resulting in controlled drug release. Drug release was slowed with an increasing amount of CNC in the PCL nanofibers.

2.2. CNFs as a Vehicle for Drug Delivery

Löbmann and Svagan [114,115,116] reported about CNFs used in drug formulations, with a focus on poorly soluble drugs. CNFs are used as stabilizers for achieving long-lasting sustained release and serves as film and foam formers.
Bhandari et al. [117] studied CNF aerogels as carriers for oral controlled drug delivery systems. Aerogels exhibited favorable floatability and mucoadhesive properties and were prepared with the lyophilization method. Paukkonen et al. [118] developed emulsion for immediate and sustained drug release applications. The emulsion was made from natural biopolymers, Class II hydrophobin protein HFBII from Trichoderma reesei and NFCs. Further, Svagan et al. [119] prepared floating solid CNF-based nanofoams loaded with furosemide. Foams exhibited sustained release of a model drug, which, in turn, depended on the drug loading, foam dimension, as well as the solid-state of the drug. Fakhri et al. [120] developed Fe3O4-Ag2O quantum dots/CNF nanocomposites and grafted them with two anticancer drugs. The results of the study showed that nanocomposites could be applied to the drug delivery system for treating skin cancer. Paukkonen et al. [121] used anionic CNF hydrogels for the delivery of small molecules and proteins. Freeze-drying into aerogels and redispersion into hydrogels of the before mentioned hydrogels is also possible. Guo and colleagues [122] fabricated CNF/alginate beads for the release of metformin hydrochloride. CNFs improved encapsulation efficiency and enabled more sustained drug release. Poonguzhali et al. [123] prepared alginate and CNF film loaded with ampicillin and investigated in vitro drug release. NC and ampicillin did improve the swelling and mechanical properties of alginate; moreover, ampicillin-loaded films exhibited good drug delivery systems with sustained drug release. Liu and colleagues [124] combined CNF and polydopamine (PDA) with calcium ion as a cross-linker. Tetracycline hydrochloride (TH) was used as a model drug for testing hydrogel as potential drug delivery carriers. In vivo skin defect experiments showed the synergistic effect of hydrogel on promoting wound healing. Sarkar, Orasugh et al. [125] prepared CNF/chitosan (CNF/CS) film with potential use in transdermal delivery. The film was loaded with an anti-inflammatory and analgesic agent, ketorolac tromethamine (KT). Drug release of KT was sustained with the incorporation of CFNs, and, therefore, this nanocarrier could be a potential candidate for transdermal drug delivery systems. Further, Orasugh et al. [126] formulated a nanocomposite of CNF and hydroxypropylmethylcellulose (HPMC). The nanocomposite was loaded with KT. Moreover, in vitro drug release results showed that the cumulative drug release rate decreased with the increase of CNF concentration in nanocomposites. An aim of Auvinen and colleagues’ [127] study was to modulate the drug release properties of CNF hydrogel. To limit the direction of drug diffusion, the first step of their study was to manufacture non-active capsules with the 3D printing method, using poly(lactic acid) (PLA). Next, drug dispersion was made of model compounds (beta-blocker metoprolol and nadolol) and anionic CNF hydrogel. The results showed that, by adjusting the geometry of the 3D printed PLA capsule, sustained release profiles provided by the CNF matrix could be modulated accurately. Importantly, however, the release of any CNF-compatible drug can be modulated easily by altering the inner geometry of the PLA capsule. Further, CNF and gelatin cryogels (hydrogels, synthesized at subzero temperatures [128]) were used for delivery of the widely used compound for treating cancer, 5-fluorouracil [129]. The structure of cryogels, CNF/gelatin ratio, density, cross-linking degree, and pH values all influence the behaviors of drug release. The duration of the sustained drug release time is up to 12 h. Meneguin et al. [130] reinforced starch/pectin free-standing films with CNFs or BNC fibers, and tested them for colonic methotrexate release. The study showed that CNFs are the best nanofiller, as the addition of CNFs improved the mucoadhesive, barrier, mechanical, and release properties of films. Such films are promising as poor solubility drugs carriers, as they increased drug dissolution rates, with approx. 80% of methotrexate release in 2 h and 30 min.
Fiorati and coworkers [131] tested the mechanical and drug release characteristics of aerogels made from cross-linked CNFs using amine-containing polymers and citric acid. Ibuprofen and amoxicillin were used as model drugs, and the results showed good absorbing properties for possible drug delivery applications. Recently, however, the same group of researchers [132] made stable hydrogels from TEMPO-CNFs with added Ca2+ ions. Proposed hydrogels are suitable for control drug delivery of ibuprofen and are proven to be cytocompatible. In particular, it is interesting to note that the addition of a certain amount of polyvalent cation can adapt the physicochemical properties of the material, and the formation of self-sustaining hydrogels is possible.

2.3. BNC as a Vehicle for Drug Delivery

BNC is a widely explored nanostructured matrix in different applications and is used in many fields, such as the food industry, drug delivery, biomedical materials, nanostructured biomaterials, and Tissue Engineering [24].
Alkhatib et al. [133] produced a BNC/poloxamer hybrid system that provides prolonged retention time for the octenidine, an antiseptic drug. An aim of the next study [134] was to develop fish scale-BNC biopolymer composite microneedles and load them with lidocaine, the medication most often used for numbing tissue in a specific location. Prepared microneedles could pierce the outermost layer of the epidermis and dissolve in the skin to release loaded drugs, in this case, lidocaine. Further, a group of researchers [135] developed pH-sensitive systems for the controlled release of diclofenac based on poly(N-methacryloyl glycine)/BNC composites, whose thermal, mechanical and viscoelastic properties were good. These nanocomposites have high water uptake capacity, are non-cytotoxic and pH-sensitive (the drug was released at pH 7.4, while at pH 2.1, the drug was retained in the nanocomposite) and, therefore, also suitable as carriers for dermal and oral administration. Ratnayake et al. [136] investigated freeze-dried BNC as a matrix for controlled protein delivery. Due to its high-water solubility, abundance, and good stability, bovine Serum Albumin (BSA) has been used as a model drug. The results of the study showed a good pattern for the loading and release profiles in the system used and, moreover, research indicates the importance of BNC as a carrier for protein drugs. Fey at al. [137] showed that BNC is suitable as a carrier for intestinal epithelial cells in drug delivery studies. Further, Silva et al. [138] investigated long-term storage of BNC membranes, loaded with lipophilic and hydrophilic active pharmaceutical ingredients (APIs). They evaluated stability at different temperatures and relative humidity. The results of the study showed that all BNC membranes loaded with APIs were stable and did not change either structurally or morphologically. Due to the hydrophilic nature of BNC membranes, the moisture-uptake increased with relative humidity. Meanwhile, Abba et al. [139] studied the incorporation of crocin (water-soluble pigmented carotenoid of saffron, which possesses antioxidant, antitumor, memory enhancer, antidepressant, anxiolytic and aphrodisiac properties [140]) into a BNC membrane. The results of direct dissolution and transdermal pass have shown compelling release, making BNC membranes a promising way for delivery of crocin.

3. Nanocellulose Based Antimicrobial Hybrids and the Use of Antimicrobials in Drug Delivery

It is important to note that native (as produced) NC does not possess antimicrobial properties. This can be achieved by functionalizing it or incorporating antimicrobial agents [55].
Halogens, phenols, silver nanoparticles and quaternary ammonium salts are used widely as antimicrobial agents [141]. Silver has been used most extensively for healing infections because its nanoparticles have proved to have antimicrobial characteristics [142]. Metal oxide nanoparticles (TiO2, CuO, ZnO, MgO) also exhibit antimicrobial activity [65]. For less toxic and more sustainable effect, some organic antimicrobial agents, such as porphyrin, lysozyme, lactoperoxidase, lactoferrin, chitosan-benzalkonium chloride, chitosan-methylisothiazolinone, gentamicin, ɛ-polylysine and sorbic acid were incorporated in NC [47,72,143,144,145]. These nanomaterials have promising antimicrobial properties against Gram-positive and Gram-negative bacteria.
An unexplored and unknown area is the balance between better antimicrobial activity, the duration of its effect and control of human cell damage [6].

3.1. Inorganic Hybrids

New therapeutic agents are also based on the use of metal nanoparticles that have an antimicrobial effect. Most commonly used and researched are silver nanoparticles (AgNPs) because of their properties such as size, shape, broad spectrum of antimicrobial activity and others [146]. Mostly they are conjugated with BNC or CNFs. They are attractive for research because AgNPs did exhibit strong cytotoxicity against fungi and viruses [147], besides quality growth inhibition of bacteria such as Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, Bacillus subtilis, Klebsiella pneumonia, Vibrio cholerae, Pseudomonas aeruginosa, and Salmonella typhi [146,148,149,150,151,152,153]. Furthermore, AgNPs can enter inside the bacteria, accumulate on bacteria cells’ enzymes and proteins and interact with deoxyribonucleic acid, which causes further damage and, consequently, bacteria inactivate as well [149].
Various methods for the preparation of AgNPs/NC hybrids have been described [23,66,154]. Mostly used is the AgNO3 solution combined with strong reducing agents. NC membrane is immersed into a silver precursor (AgNO3), so agglomerates are immobilized only by physical interactions [155]. The schematic procedure of the preparation of BNC-AgNPs hybrids is shown in Figure 5.
The described hybrids have proved good antimicrobial activity against B. subtilis, E. coli, and S. aureus [23,65,156]. Furthermore, loading BNC membranes with Ag nanoparticles showed 99% growth inhibition of E. coli, S. aureus and P. aeruginosa [157]. The antimicrobial properties have been proven, so the connection between silver and drug delivery is interesting. Incorporation of silver ions within biocompatible and osteoconductive biomaterial hydroxyapatite (HAp) has been performed in many studies, with the option of long term silver ion release rates [158,159,160]. Furthermore, AgNPs were previously incorporated in chitosan hydrogels [161] and hybridized structures [162], thereby achieving prolonged and controlled release. The drug release rate and hydrophilic ratio also increased-with-increased Ag concentration.
As the biocompatibility of AgNPs has not yet been determined, and it is well known that AgNPs’ toxicity is concentration-dependent, the use of such hybrids in the field of Biomedicine is limited [163,164]. Therefore, Lizundia et al. [165] designed antimicrobial bio-based films composed of CNCs and metallic (Ag, ZnO, TiO2) nanoparticles. They all showed antibacterial effectiveness against E. coli and S. aureus.
Many metal oxide nanoparticles have already proven successful in drug delivery systems [166]. For example, as a type of pH-responsive drug carrier, ZnO nanostructured materials were suggested in 2010 [167,168]. TiO2 particles have also been used for such drug carriers [169], and later these materials were often applied for drug delivery systems [170]. The beforementioned and some other studies of inorganic NC-based antimicrobial hybrids are summarized in Table 4.

3.2. Organic Hybrids

Natural polymers such as polysaccharides and proteins are becoming an important basis for the development of antimicrobial materials for various applications. While some biopolymers (e.g., chitosan and lysozyme) are natural biocides, others (e.g., cellulose) must be bind to bioactive compounds to obtain such properties [179].
Among many applications (e.g., reinforcing agents in nanocomposites, biodegradable films, barriers for packaging, stabilizing agents in dispersions for technical films and membranes, additives in food, texturing agents in cosmetics [180]), CNFs have been used to prepare thin films for wound dressing and bioactive implants [181]. Most NC applications in the Biomedical field are still progressing in the laboratory, assessing its ability, capacity, reproducibility, and effectiveness in providing its roles as scaffolding and regeneration, pharmaceutical applications (e.g., drug carrier) and implants [182,183,184,185].
The biggest problem with the wound healing process is a secondary infection, where bacterial sepsis can be fatal in the case of some extreme burns. Because NC itself has no antimicrobial properties, it needs to be modified [186], e.g., loading the BNC membrane with an antimicrobial agent such as benzalkonium chloride (BZC) [187] or polyhexanide (PHMB) and povidone-iodine (PI) [188]. BZC is an antimicrobial cationic surfactant, which was used widely in commercial wound dressings. It effectively inhibited the growth of various bacteria (E. coli, S. typhimurium, B. subtilis). Previously, BZC has already been used in drug delivery studies [189,190,191]. For example, Garcia-Fernandez et al. [192] prepared cyclodextrin-functionalized hydrogels and gauzes with BZC that could be used for the prevention and management of wound infections. PI has a high molar mass and, therefore, has a delayed-release compared to PHMB. Its important feature is good biocompatibility; however, it showed less inhibition of S. aureus growth than BNCs with PHMB. Finally, BNC loaded with PHMB provides a better and wider therapeutic window.
The next solution could be to fabricate composites of BNC with another material (e.g., chitosan [193]) showing antibacterial properties. Wiegand et al. [188] found PHMB-loaded BNCs to be promising wound dressings exhibiting controlled drug delivery, whereas PI showed significantly delayed-release compared to PHMB.
A recent study aimed to produce and characterize an optically transparent film with good antimicrobial properties from ginger CNF, fabricated using chemicals and ultrasonication. Antimicrobial effectiveness was tested using the agar disc diffusion method. Tested microorganisms in the study were S. aureus, E. coli, P. aeruginosa, B. subtilis, and C. albicans. Chloramphenicol and nystatin were used as positive controls. They tested raw ginger fibers and both cellulose film made from ginger with chemicals (alkalization, bleaching, acid hydrolysis) and with ultrasonication. All of the samples inhibited the growth of all microorganisms. Researchers concluded that the sonication process does not damage the fiber’s natural antimicrobial properties because there was no perceptible difference in antimicrobial effectiveness between sonicated and acid hydrolyzed nanofiber films [194].
On the other hand, some studies used different ginger-derived substances (lipids [195], phenolic compounds [196,197]) in drug delivery systems. Zhang et al. [195] describe a ginger-derived nanovector made of ginger-derived lipids that can serve as a delivery platform for the therapeutic agent to treat colon cancer. In a subsequent study, Naghs [197] described the potential anti-inflammatory and anticancer action of ginger extracts jointed with nanotubes as a technique for drug delivery.
Chitosan (CS), which is derived from chitin (Figure 6) and is one of the naturally-occurring antimicrobial agents, has gained much popularity in commercial applications [198].
Due to its molecular weight, degree of deacetylation, positive charge and mucoadhesive nature [200,201,202], it is considered an efficient matrix molecule or carrier in drug delivery systems [203,204,205,206,207,208]. Precisely because of the increase in bioavailability, mucoadhesive substances (e.g., chitosan, cellulose derivatives [209]) are usually used in pharmaceutical formulations, which have an increased affinity for mucosal membranes and adhere to mucous membranes. With carefully planned mucoadhesion, specific retention of the active substance at a particular site is possible, and a controlled rate of drug release can be achieved [81].
Chitosan and its derivatives have been used widely in many biomedical applications [210], including in antimicrobial wound dressing [211], drug delivery systems [212], and tissue engineering [213].
Already in 1998, Felt and her colleagues [214] wrote of chitosan as a unique polysaccharide for drug delivery. Their article describes many applications of chitosan in the oral, injectable, nasal, ophthalmic, and transdermal routes of administration. More recent studies of drug delivery applications of chitosan and chitosan-based NPs are described in reviews from Pahri [215] and Naskar et al. [216].
By adding chitosan to NC (Figure 7), the researchers improved the mechanical properties of the NC and contributed to the antimicrobial effectiveness [217] of the cellulose-based material. Chitosan is a naturally occurring amino polysaccharide with outstanding properties such as biodegradability, biocompatibility, nontoxicity, healing enhancement, and, especially, antibacterial properties [218,219].
Researchers studied the antibacterial effectiveness of hydrogel CS/BNC and lyophilized CS/BNC against E. coli and S. aureus. For the lyophilized samples, the number of bacterial colonies was significantly lower than the BNC control group. For the hydrogel samples, the bacterial numbers on all CS/BNC composites were lower than the BNC group but higher than the number just after inoculation, also showing a bacteriostatic ability. The antibacterial effect of lyophilized samples was much stronger than that of the hydrogel samples. These properties provide fabric-reinforced CS/BNC composites with great potential as excellent medical materials for wound dressings [220].
For comparison, the following study covers the use of CNCs in the synthesis of antimicrobial hybrids with chitosan. Poonguzhali et al. [222] studied chitosan polyvinylpyrrolidone (PVP)-NC (CPN) bionanocomposites for wound dressing application. PVP, polyvinyl pyrrolidone, is a non-toxic, biocompatible polymer usually applied in controlled drug release and wound dressing. It exhibits hydrophilic properties, universal solubility, and a good tendency for the formation of complexes with a wide range of molecules. Due to the latter properties, these polymeric films are attractive drug delivery systems, especially for systemic effect through the sublingual and buccal routes [223,224,225]. CPN was tested against S. aureus, and P. aeruginosa strains with modified agar diffusion assay. The largest inhibition zones were observed for the Gram-negative bacteria P. aeruginosa, while smaller was observed for the Gram-positive S. aureus. They suggest that the reason for better inhibition of Gram-negative bacteria may be that chitosan is charged positively and microbial cell membranes are charged negatively. Therefore, the interaction between them leads to bacterial leakage by disrupting the bacterial strain’s metabolism. In addition, the extremely large NC surface eased the adsorption of the target bacteria, which accelerated antimicrobial efficiency.
Furthermore, Hasan et al. [226] prepared composites from chitosan, PVP and CNCs for delivery and release of curcumin in wound dressing application. CNCs improved the thermal, swelling, and mechanical properties of the film. In addition, better protein absorption was observed in the presence of CNCs in the films, which improved the wound healing properties of the dressing material. The proposed release system is a potential dressing material in wound healing applications.
Recently, antimicrobial tissue paper’s potential for preventing the spread of infectious diseases has been developed. Potential inhibition of the spread of COVID-19 has also been reported [227,228], but these claims are not substantiated. To confirm these speculations, antimicrobial tissue paper should be tested specifically against COVID-19 to determine the effectiveness of these materials. Researchers prepared hand-sheets which have been spray-coated with chitosan, CNC, and their composite coating (chitosan-CNC). The main purpose of the study from Tyagi et al. [229] was to empower tissue paper composites with a synergistic effect of CNC and chitosan on increased water absorbency, mechanical strength, and antimicrobial activity. Tissue paper sheets were made with the use of a handsheet mold from recycled, bleached, deinked pulp. Chitosan solution was prepared in 1% acetic acid, which was further mixed with CNC for 1 h at 500 rpm. The ratio of chitosan/CNC was 80:20 by weight. Before surface coating, researchers conditioned the handsheets at 50% relative humidity and 23 °C for 24 h. chitosan-CNC coatings (1% solid concentration) were spray-coated on the handsheets and further dried at 110 °C. The preparation of such antimicrobial tissue papers is depicted in Figure 8.
Furthermore, tissues exhibited antimicrobial activity against E. coli, the growth of which was inhibited by up to 98%. chitosan-CNC coatings exhibited a zone of inhibition with a diameter of 17.4 mm. The synergistic effect of chitosan and CNC proved to be excellent, as coatings with chitosan alone showed an 8.7 mm zone of inhibition. The coatings were also improved by plasma treatment, as the plasma-treated chitosan-CNC coated tissue paper exhibited the highest zone of inhibition, with a diameter of 23.4 mm and a 99% reduction in the growth of E. coli. Plasma-treated coatings additionally showed antimicrobial activity against microbes from a human–hand sample, in which the presence of many different bacteria, fungi and virus is expected. The atmospheric plasma used was composed of helium as an inert gas and oxygen as a reactive gas, and certainly, oxygen plasma is known to increase the hydrophilic nature of tissue paper, resulting in increased antimicrobial activity of plasma-treated chitosan-CNC coated tissue paper [227,228,229,230].
Not only BNC and CNCs, but CNFs are also used in combination with chitosan and other active ingredients to achieve an antimicrobial effect. For packaging applications, Sundaram et al. [231] developed biodegradable composite membranes with antimicrobial properties. They combined CNFs, chitosan and S-nitroso-N-acetyl-D-penicillamine (SNAP). The zone of inhibition showed against S. aureus, E. faecalis, and L. monocytogenes. The point is that the SNAP molecule contains a group of nitric oxide (NO), which inhibits the growth of bacteria (both Gram-positive and Gram-negative), viruses, fungi, and yeast [232]. In addition, it is an endogenous antiplatelet agent, and therefore the exogenous release of NO from different polymer matrices has shown the reduction of thrombosis and infection of implantable medical devices [233].
A recent study examined the antimicrobial effectiveness of NC and grape seed extract added chitosan and polycaprolactone (PCL) based biofilms [234]. Grape seed extract contains many phenolics, such as catechins, epicatechin, gallic acid, and procyanidins. These have shown various biological effects, including antimicrobial abilities [235]. Further, PCL is an aliphatic polyester, and when mixed with chitosan, allows it to maintain its mechanical properties and improve its barrier properties. Films like that could be used in food packaging applications. All film samples showed antimicrobial activity against E. coli and Listeria monocytogenes. The high permeability of PCL caused by its rubbery characteristics enables the delivery of low molecular weight drugs such as vaccines [236] and steroids [237]. Compared to other biodegradable polyesters, PCL has become interesting in drug delivery due to its lack of toxicity and low cost. However, due to its high crystallinity and hydrophobicity, its deficiency is a slow degradation rate in vivo.
The solution, however, is that PCL biodegradability can be increased by copolymerization or by blending it with other polymers. Therefore, Sahoo et al. [238] blended chitosan with PCL for controlled delivery of ofloxacin. In addition to all the other good properties of chitosan (e.g., low cost, availability, positive charge, biocompatibility, and antimicrobial activity), this blend is a more biodegradable and biocompatible material that can be used in controlled drug delivery systems.
The production of antimicrobial NC membranes can be achieved by chemical grafting of functional groups on the surface of cellulose nanofibers (e.g., CNF functionalized with amino and aminosilane groups, which inhibited the growth of E. coli and S. aureus) [141,239,240].
For example, a chemical grafting of aminoalkyl groups onto the surface of BNC membranes may be used to mimic the internal antimicrobial properties of chitosan. The presence of free amino groups along the polymer chain is responsible for the antimicrobial efficacy of chitosan.
Fernandes and Sadocco et al. [73] used 3-aminopropyltrimethoxysilan (ASP) to achieve chemically grafted aminoalkyl groups at the surface of BNC (Figure 9). They tested antimicrobial activity against S. aureus and E. coli strains using a standard dynamic shake flask method. BNC-NH2 membranes showed a significant reduction in bacterial viability for both E. coli and S. aureus. A study showed that the silane chemical grafting approach produces a BNC membrane with antimicrobial activity while maintaining biocompatibility.
Furthermore, BNC is a highly pure form of cellulose and a swollen membrane with high water content [241]. However, due to its physical and mechanical properties, this biopolymer is of great interest for use in the Biomedical field, and BNC-NH2 membranes have potential for applications in tissue engineering, wound healing, and drug delivery systems.
The purpose of the following study was to evaluate the antimicrobial activity of BNCs filled with nisin. Nisin is a 34-amino acid long bacteriocin, which is active against many foodborne bacteria [242]. BNC membranes were immersed into a nisin solution with or without EDTA, kept in a shaker and then immersed in phosphate buffer solution (PBS). The antimicrobial activity of nisin, whether using EDTA solution or not, was assessed by minimal inhibitory concentration (MIC) and the agar diffusion method. Tested microorganisms were S. aureus, E. coli, and P. aeruginosa. A combination of nisin solution with EDTA showed a synergistic effect. For E. coli and S. aureus, antimicrobial activity was observed, while for P. aeruginosa, it was not. BNC membranes loaded with nisin are promising agents to prevent microorganism contamination [243].
The delivery of nisin for antimicrobial efficacy has been investigated extensively in recent years. Ugurlu et al. [244] found out that an envelope from pectin and hydroxypropyl methylcellulose (HPMC) is a good delivery system for nisin to be delivered to the colon. Further, Coelho Correia et al. [245] found a poly(lactic-co-glycolic acid) (PLGA)-nisin matrix promising for sustained drug delivery, with continuous release of nisin for two weeks. Nisin embedded in polymer matrices has, thus, the potential for topical drug delivery. Shin and colleagues [246] described various biomedical applications and delivery of nisin, including the treatment of various infections, mastitis, oral health, and cancer.
Further, BNC was loaded with bromelain (BL). That is a protease found in pineapple tissues [247,248]. bromelain was incorporated in BNC membranes by immerging membranes in a bromelain solution. The bromelain antimicrobial activity is related to its enzymatic activity. They tested the initial bromelain solution, residual bromelain solution, and bromelain solution after being released from BNCs. Results showed that it inhibited all tested microorganisms (E. coli, S. aureus, and P. aeruginosa), especially for the release solution.
In addition, the BNC-BL membrane proved to be an auspicious drug delivery system that shortens healing time, reduces the risk of infections, and helps relieve pain [249]. Bagga et al. [250] used bromelain capped gold NPs as a drug delivery carrier of the antibiotic levofloxacin. Due mainly to the stability of such NPs and the delivery of a large number of levofloxacin molecules to a highly localized area at the point of contact between the NPs and the bacteria, the NPs have proven as effective carriers of the selected antibiotic and, further, improved antimicrobial activity against Gram-positive and Gram-negative bacteria. BL is known as a phytotherapeutic anticancer agent [251], but its activity is decreased upon oral administration, and for this reason, Bhatnagar et al. [252] encapsulated BL into PLGA to formulate NPs and coated them with polymer for stability against acidic gastric conditions. Based on the generation of reactive oxygen species, induction of apoptosis and weakened mitochondrial membrane potential in Ehrlich cells (in mice), these NPs are potential candidates for oral chemotherapy.
Enzybiotics are particularly interesting. They have been proclaimed as an environmentally safe and interesting alternative to antibiotics as antimicrobial agents. Today, all enzymes with antibacterial and antifungal properties are included in the enzybiotic group [253].
Antimicrobial activity of crude laccase against Gram-positive and Gram-negative bacteria has been observed, which is interesting because laccase is known to catalyze reactions, leading to the emergence of antimicrobial species. BNC membranes were lyophilized and immersed in the laccase preparation diluted in a phosphate-citrate buffer. The antimicrobial effect was evaluated against S. aureus and E. coli. Antimicrobial activity was compared with Ag nanoparticles (AgNPs), and Laccase/AgNPs immobilized on BNC. The results showed the antimicrobial effect of the laccase. Moreover, laccase inhibited the growth of Gram-positive bacteria better than Gram-negative ones (about 92% (S. aureus) and 26% (E. coli) of bacterial inhibition) [254].
Lacasse was not observed in literature for drug delivery systems, but recently, Zhang and colleagues [255] revealed a food gel of soluble crosslinked corn bran arabinoxylan (CAX). Alkali-extracted CAX was treated with laccase to form soluble crosslinked CAX, which formed a gel on pH reduction. Such gels could be taken in the low pH environment of the stomach (e.g., as food gels or beverages containing soluble crosslinked CAX) and as a drug delivery matrix.
After a few listed applications with BNC, the following study [108] used CNC film as a wound dressing material and conjugated it with curcumin. It is a lipophilic phytopolyphenol isolated from Curcuma longa. With the controlled release of curcumin, they wanted to achieve a long-lasting antimicrobial effect of the NC film. They used the agar diffusion test and Hohenstein challenge test (AATCC-100) as indicators for antimicrobial activity. The results of the study were great, as curcumin loaded film showed significant antimicrobial activity on three Gram-positive (methicillin-resistant S. aureus, Streptococcus sp., Bacillus coagulans), two Gram-negative (E. coli, Proteus mirabilis) bacteria and one yeast (C. albicans). Curcumin-loaded film reduced the growth of all tested microbial cells significantly, as five of six test microorganisms showed a 99% growth reduction relative to the control.
Furthermore, Raghavendra et al. [256] impregnated nanocurcumin into gelatin nanocellulose fibers for potential antimicrobial activity and further applications. Study results showed antimicrobial activity against E. coli and S. aureus for 24 h. Moreover, curcumin possesses chemopreventive, chemotherapeutic, antioxidant, anti-inflammatory and hyperlipidemic properties [257].
Due to poor aqueous solubility and poor oral bioavailability, the therapeutic efficacy of curcumin is limited [258], but it is still used widely in applications for drug delivery systems because of its outstanding antimicrobial properties. For example, Sanoj Rejinold et al. [259] formulated highly stable thermoresponsive nano constructs of curcumin with chitosan-g-poly (N-isopropylacrylamide) for drug delivery, Sun et al. [260] used exosomes, released from a number of different cell types, as a vehicle for curcumin delivery, and Pham et al. [261] modified curcumin-loaded superparamagnetic iron oxide NPs with chitosan for use as drug delivery carriers in the treatment of cancer cells. Recently, Gupta et al. [262] published a review of the latest curcumin-based nanoformulations for targeting different diseases.
Improving the properties of CNCs and adding antimicrobial activity to them can be done by esterification of the surface of CNCs using non-toxic resin acids, rosin (colophony; a natural product of coniferous trees [263]). This was proven by de Castro et al. [264]. The rosin-grafted CNC showed strong antibacterial effectiveness against Gram-negative bacteria (E. coli) and small antibacterial activity against Gram-positive bacteria (B. subtilis).
Rosin is one of the highly stable, non-toxic, biodegradable, and gel-forming natural gums. It possesses film-forming and coating ability, and further, it has been used as microencapsulating agents and as anhydrous binders, or matrix material in tablets for controlled drug release [265]. Kumar Yadav et al. [266] described and reviewed pharmaceutical applications of rosin, including film-forming materials, transdermal drug delivery, and targeted drug delivery. It was used mainly as a taste-making agent, microencapsulating agent and material, binding agent, matrix-forming material, and emulsifying agent for achieving good stability, faster dissolving and sustained controlled, or sustained drug release.
Preparation and investigation of antibacterial materials by utilizing protein degrading enzymes (e.g., lysozyme) is also interesting. Such enzymes degrade bacteria by adherence [267].
Lysozyme is antimicrobially effective against Gram-positive and Gram-negative bacteria [268]. Uddin, Orlema et al. [267] investigated the antibacterial activity of CNF-lysozyme aerogels and compared them with silver-containing CNF aerogels. Lysozyme aerogels showed antibacterial effect at E. coli and S. aureus strains, as did silver CNF aerogels, which antibacterial activity depended on the nanoparticle concentration. It was observed that the higher the Ag nanoparticle concentration, the higher the inhibition of the microbial culture.
Besides antimicrobial activity, lysozyme exhibits antitumor, antiviral and immune-modulatory activities; therefore, having the ability to suppress tumor cells [269]. Alves et al. [270] studied the potential use of fluorinated ionic liquids (FILs) as a drug delivery system for proteins. They used lysozyme as a model protein and investigated the impact of FILs on its structure and function. Its hydrolytic performance, therefore, its stability and activity did not decrease. Further, Wu et al. [271] prepared chitosan/sodium alginate-based (CS-ALG) hydrogels for the delivery of lysozyme. Antibacterial activity of hydrogels was improved with the addition of lysozyme, and the relative activity of the released lysozyme was 87.72 % ± 3.96%, which makes CS-ALG hydrogels a promising matrix for enzyme loading and their delivery.
Allicin is an antimicrobial agent that forms when a clove of garlic is freshly crushed. It is formed when the enzyme alliinase gets in contact with alliin and converts it to allicin [272]. Jamindar et al. [273] formulated and characterized allicin-amphotericin-B liposomal gel for topical treatment of fungal infections, but a recent study from Jebali and coworkers [274] describes the antimicrobial activity of cellulose NPs conjugated with allicin and lysozyme separately (Figure 10).
Antimicrobial properties were determined with the microdilution method and compared to allicin, lysozyme, and CNC alone. The allicin and lysozyme were conjugated to CNC by a carbodiimide cross-linker, both showing good antifungal and antibacterial effects against Aspergillus niger, C. albicans, S. aureus, and E. coli [274,275].
Atef et al. [276] incorporated summer savory (Satureja hortensis L. [277]) essential oil into agar-based nanocellulose films. They evaluated antimicrobial activity with the disc diffusion method against L. monocytogenes, S. aureus, B. cereus, and E. coli. Samples without savory essential oil (SEO) were considered as control and did not exhibit any zone of inhibition. The addition of SEO showed a weak inhibitory effect. L. monocytogenes and B. cereus were the most sensitive microorganisms to the SEO containing nanofilms, and E. coli was the most resistant bacterium. As for delivery systems, SEO was not used, except when Cansu Feyzioglu and Tornuk [278] formulated chitosan NPs loaded with SEO for potential antioxidant and antimicrobial delivery applications.
The next few antimicrobials were conjugated with cellulose and also have great potential for conjugation with NC materials. As a result, they could be good NC-based antimicrobial hybrids, acting as an alternative to classic antimicrobials.
One of the natural antibiotics is propolis. Conjugated with BC, it shows potent antimicrobial activity and wound healing properties [279,280]. Ethanol extracts of propolis can inhibit the growth of S. aureus, Enterococcus sp. and P. aeruginosa [281].
Propolis has been involved widely in research on drug delivery systems. In 2010 Dantas et al. [282] developed propolis microemulsion for topical applications. Ahmed et al. [283] found out that Egyptian propolis extract in a cold cream formulation, alone or in combination with Dermazine, is appropriate for topical administration for the treatment of diabetic burn wounds. Further, Berretta et al. [284] created a formulation with 3.6% propolis extract, which can benefit in the treatment of skin injuries. Balata et al. [285] formulated pluronic lecithin organogel with propolis that exhibited great skin permeation and antimicrobial activity, and later, Rassu et al. [286] prepared propolis as lipid bioactive nanocarrier. Propolis-based solid lipid NPs could be used as delivery systems of drug and flavonoids and for the local treatment of nasal diseases.
Honey from the Manuka tree conjugated with BC also showed antibacterial activity. Its quality is to promote tissue growth, so Manuka honey is one of the promising agents in wound healing applications [287]. Drug delivery dressings are defined as substances applied directly to the surface of the wound to display therapeutic properties in terms of benefit to the healing process. Manuka honey could be a therapeutic agent and vehicle itself and can be compared to many other natural therapeutic polymers (e.g., alginate, chitosan, collagen, and hyaluronic acid) used as drug delivery dressings [288,289]. Tenci et al. [290] developed a powder formulation for delivery of manuka honey bioactive components and platelet lysate (PL) in chronic skin ulcers.
Cinnamaldehyde and eugenol (the main ingredients of cinnamon [291] and cloves [292]) were incorporated with cellulose films to obtain active antimicrobial materials. Antimicrobial effectiveness was investigated against nine bacteria (B. cereus, E. faecalis, L. monocytogenes, Micrococcus luteus, S. aureus, Aeromonas hydrophila, E. coli, P. aeruginosa, Salmonella enteritidis) and three strains of yeast (C. albicans, Saccharomyces cerevisiae, Zygosaccharomyces rouxii). The agar well diffusion method and a vapor diffusion technique were used for its determination. A. hydrophila and E. faecalis were the most sensitive to cinnamaldehyde or eugenol films. The cellulose-based film containing cinnamaldehyde or eugenol did not completely inhibit the growth of microbial cells. However, they showed positive activity against all selected strains in terms of size and number of microbial colonies in the vapor diffusion test. This study demonstrates the potential use of cinnamaldehyde and eugenol for application to an antimicrobial film or coating [293].
Recently, Kenawy et al. [294] combined gelatin, chitosan, and cinnamaldehyde and developed biodegradable crosslinked membranes that demonstrate the potential as antimicrobial dressings for mitigating wound healing, and, interestingly, Bang and Kim [295] developed a stable self-microemulsifying drug delivery system for trans-cinnamaldehyde use in the Pharmaceutical industry.
Also, eugenol is used widely in drug delivery systems. Patole et al. [296] used it with thymol in loaded chitosan dental film for the treatment of periodontitis, Esmaeili et al. [297] studied the effects of eugenol nanoemulsion as a topical delivery system, and Khayat et al. [298] developed eugenol microemulsion for transdermal delivery of drugs.
Aloe vera has great potential for biomedical applications because of its biological activity. For optimized properties, aloe vera has been added to chitosan in previous studies [299], but recently, researchers reported the production of aloe vera and bacterial cellulose composites [300,301]. No NC-based antimicrobial studies were detected in the literature reviewed.
Gupta et al. [302] investigated the antimicrobial activity of aloe vera loaded poly(vinyl alcohol) (PVA)/poly(ethylene oxide) (PEO)/carboxymethyl cellulose (CMC) coated polyester membranes. They evaluated antimicrobial properties against S. aureus and E. coli. The number of viable cells decreased in the membrane containing aloe vera as compared to a non-aloe vera control sample. aloe vera has been found to be effective against both Gram-positive and Gram-negative bacteria.
In future studies, it would be interesting to incorporate aloe vera into NC materials and to check their antimicrobial activity and efficacy. Laux and coworkers [303] claim that aloe vera gel and whole leaf could be used as an excipient in drug delivery systems. Their article is a review of the role of aloe vera leaf materials in oral, transdermal, and buccal drug delivery. Such materials could have good potential in various biomedical applications.
The beforementioned and some other studies of organic NC-based antimicrobial hybrids are summarized in Table 5.

4. Challenges and Future Prospects

NC extraction methods, raw material selection, environmental and biological toxicity, and life cycle assessment are extremely important for the development of sustainable applications in which NC has strong potential. NC can be extracted using different techniques from many different sources. In order to optimize the process and adapt the properties and characteristics of NC, it is important that future development include mainly sustainable, eco-friendly, and green isolation techniques, such as mechanical extractions and enzymatic hydrolysis looks the most promising. Most of the studies using advanced technology were successfully performed on a laboratory scale, and further research activities need to be conducted to transfer them to the industrial scale. Due to commercialization, it is necessary to look at the whole aspect and understand the impact on the environment that encompasses the life cycle of NC, as only this can enable its sustainable success. Consequently, the life cycle assessment of some environmental aspects of NC based materials should be done.
BNC is especially interesting, but unfortunately, due to shortcomings of scaling up BNC production to the industrial level, which is related to its current production costs due to the relatively expensive commonly used culture media and slow production processes, is this replacement currently less feasible.
In addition, not only wood biomass is of interest for obtaining NC, but also other plant biomass, which gives cleaner and morphologically/crystallinely different products, which are of particular interest in the pharmaceutical industry and biomedicine.
The development of materials science is also advancing rapidly in the biomedical field, so nanocellulosic materials may represent a promising solution in the future to overcome some of the insurmountable challenges of biomedical materials. Much of the emphasis in this review is on the use of NC in drug delivery. Manly, the physicochemical properties and biocompatibility of NC enable its usefulness for the controlled release of various model drugs in nearly all cases. The particularly encouraging is its compatibility with both hydrophilic, hydrophobic, water-soluble, and poorly water-soluble drugs. NC is rapidly experiencing its breakthrough into drug delivery systems, but further studies are required to completely address the environment and biological toxicity of NC-based materials (to microorganisms, animals, and humans). In that respect, the assessment of the potential risks attributed to the chemical modification of nanocellulosic material requires special attention.
Because many NC-based hybrids containing biopolymers, surfactants, bacteriocins, enzybiotics, phytophenols and other organic compounds have shown antimicrobial efficacy in the studies reviewed, the potential in this area is extremely high. Hence, the future lies mainly in natural extracts combined with NC (biomimetically structured materials), on which extremely little research has been done. Natural extracts, with their biocompatibility and nontoxicity, enable many applications. Given that natural extracts do not consist of only one component but contain several different ones, they can act synergistically. Thus, in vitro and in vivo studies are needed to predict the synergistic effect of natural extracts on organisms and to allow the breakthrough of such NC hybrids to actual use, especially in biomedicine.
Especially highlighting is also focused on the food industry, where the demand for new and innovative food packaging materials is growing. In particular, antimicrobial bionanocomposites are promising for the packaging of various problematic foods (e.g., meat products, dairy and bakery products, fruits and vegetables), where spoilage occurs due to microorganisms. A new packaging technology (active packaging) incorporates natural extracts into NC-based films to control microbial surface contamination of foods.
With the realization of the above-mentioned challenges, NC-based materials may certainly contribute to the improved quality of human life in the future, especially through the development of the next generation of NC hybrid materials.

5. Conclusions

Over the last decade, there has been an increasing number of research teams around the world who reported the use of nanocellulose/NC in drug delivery studies and modification of NC with different antimicrobial agents. This review aims to outline the current state of research and the future development of NC in antimicrobial and drug delivery applications through selected examples.
Most formulations of NC-based drugs can be used through various routes of administration and have demonstrated controlled and, in many cases, also sustained drug release due to the addition of NC. Moreover, NC shows tremendous potential for further study and even more successful use in controlled drug delivery systems.
The surface-modified antimicrobial films from NC present a wide range of possibilities in applications in the areas of medicine (e.g., wound healing), pharmacy (e.g., drug carriers), food packaging, etc., NC-based nanomaterials incorporated with both inorganic and organic antimicrobial agents showed extremely good antibacterial activity against both Gram-positive and Gram-negative bacteria. Some of the specifically functionalized NC materials also provide antifungal properties. In particular, NC materials with organic antimicrobials, because of their non-toxic and non-irritant properties, could be used as an alternative to ordinary inorganic antimicrobial agents with low molecular weight, whose bad features are potential toxicity and short-term antimicrobial efficacy. The mechanical properties of NC-based antimicrobial materials could be enhanced further. Above all, more environmentally friendly and biocompatible alternatives could be prepared since some natural materials provide good antimicrobial efficacy. NC-antimicrobial hybrids obtained from natural materials have great potential and could be used much more widely in biomedical applications.
Last but not least, NC-based antimicrobial tissues with incorporated nanoparticles can be very effective in fighting microorganisms and, consequently, infectious diseases. Overcoming antimicrobial resistance is necessary, as is the further development of new antimicrobial hybrids based on NC.

Author Contributions

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

Funding

This research was supported by the Slovenian Research Agency (ARRS) within the frame of program P2-0046 (Separation Processes and Production Design), program P2-0118 (Textile chemistry), and young researcher ARRS fellowship Contract No. 2187/FS-2019.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dufresne, A. Nanocellulose: A new ageless bionanomaterial. Mater. Today 2013, 16, 220–227. [Google Scholar] [CrossRef]
  2. Salas, C.; Nypelö, T.; Rodriguez-Abreu, C.; Carrillo, C.; Rojas, O.J. Nanocellulose properties and applications in colloids and interfaces. Curr. Opin. Colloid Interface Sci. 2014, 19, 383–396. [Google Scholar] [CrossRef]
  3. Moran, J.I.; Alvarez, V.A.; Cyras, V.P.; Vazquez, A. Extraction of cellulose and preparation of nanocellulose from sisal fibers. Cellulose 2008, 15, 149–159. [Google Scholar] [CrossRef]
  4. Muñoz-Bonilla, A.; Echeverria, C.; Sonseca, Á.; Arrieta, M.P.; Fernández-García, M. Bio-Based Polymers with Antimicrobial Properties towards Sustainable Development. Materials 2019, 12, 641. [Google Scholar] [CrossRef] [Green Version]
  5. Klemm, D.; Kramer, F.; Moritz, S.; Lindström, T.; Ankerfors, M.; Gray, D.; Dorris, A. Nanocelluloses: A New Family of Nature-Based Materials. Angew. Chem. Int. Ed. 2011, 50, 5438–5466. [Google Scholar] [CrossRef]
  6. Lin, N.; Dufresne, A. Nanocellulose in biomedicine: Current status and future prospect. Eur. Polym. J. 2014, 59, 302–325. [Google Scholar] [CrossRef] [Green Version]
  7. Bacakova, L.; Pajorova, J.; Bacakova, M.; Skogberg, A.; Kallio, P.; Kolarova, K.; Svorcik, V. Nanocellulose in Biotechnology and Medicine: Focus on Skin Tissue Engineering and Wound Healing. Preprints 2018. [Google Scholar] [CrossRef]
  8. Torres, F.G.; Troncoso, O.P.; Lopez, D.; Grande, C.; Gomez, C.M. Reversible stress softening and stress recovery of cellulose networks. Soft Matter 2009, 5, 4185–4190. [Google Scholar] [CrossRef]
  9. Habibi, Y.; Goffin, A.-L.; Schiltz, N.; Duquesne, E.; Dubois, P.; Dufresne, A. Bionanocomposites based on poly(ε-caprolactone)-grafted cellulose nanocrystals by ring-opening polymerization. J. Mater. Chem. 2008, 18, 5002–5010. [Google Scholar] [CrossRef]
  10. Wang, Y.; Cheng, Z.; Liu, Z.; Kang, H.; Liu, Y. Cellulose nanofibers/polyurethane shape memory composites with fast water-responsivity. J. Mater. Chem. B 2018, 6, 1668–1677. [Google Scholar] [CrossRef]
  11. Tree. Available online: https://arbordayblog.org/landscapedesign/12-fast-growing-shade-trees/ (accessed on 17 April 2020).
  12. Cells in Microscopic Detail. Available online: https://66.media.tumblr.com/tumblr_maqu0spvna1qc0noyo1_1280.jpg (accessed on 17 April 2020).
  13. Styela Clava-Clubbed Tunicate. Available online: http://www.marinespecies.org/photogallery.php?album=669&pic=55030 (accessed on 17 April 2020).
  14. Cladophora Sericea-Silky Green Branched Weed. Available online: http://www.aphotomarine.com/green_seaweed_cladophora_sericea.html (accessed on 17 April 2020).
  15. Xu, X.; Liu, F.; Jiang, L.; Zhu, J.Y.; Haagenson, D.; Wiesenborn, D.P. Cellulose Nanocrystals vs. Cellulose Nanofibrils: A Comparative Study on Their Microstructures and Effects as Polymer Reinforcing Agents. ACS Appl. Mater. Interfaces 2013, 5, 2999–3009. [Google Scholar] [CrossRef]
  16. Mandal, A.; Chakrabarty, D. Isolation of nanocellulose from waste sugarcane bagasse (SCB) and its characterization. Carbohydr. Polym. 2011, 86, 1291–1299. [Google Scholar] [CrossRef]
  17. Abitbol, T.; Rivkin, A.; Cao, Y.; Nevo, Y.; Abraham, E.; Ben-Shalom, T.; Lapidot, S.; Shoseyov, O. Nanocellulose, a tiny fiber with huge applications. Curr. Opin. Biotechnol. 2016, 39, 76–88. [Google Scholar] [CrossRef]
  18. Pierre, G.; Punta, C.; Delattre, C.; Melone, L.; Dubessay, P.; Fiorati, A.; Pastori, N.; Galante, Y.M.; Michaud, P. TEMPO-mediated oxidation of polysaccharides: An ongoing story. Carbohydr. Polym. 2017, 165, 71–85. [Google Scholar] [CrossRef] [Green Version]
  19. Menon, M.P.; Selvakumar, R.; Kumar, P.S.; Ramakrishna, S. Extraction and modification of cellulose nanofibers derived from biomass for environmental application. RSC Adv. 2017, 7, 42750–42773. [Google Scholar] [CrossRef] [Green Version]
  20. Shahi, N.; Min, B.; Sapkota, B.; Rangari, V.K. Eco-Friendly Cellulose Nanofiber Extraction from Sugarcane Bagasse and Film Fabrication. Sustainability 2020, 12, 6015. [Google Scholar] [CrossRef]
  21. Sharma, C.; Bhardwaj, N.K. Bacterial nanocellulose: Present status, biomedical applications and future perspectives. Mater. Sci. Eng. C-Mater. Biol. Appl. 2019, 104, 109963. [Google Scholar] [CrossRef]
  22. Khosravi, K.; Koller, M.; Akramzadeh, N.; Mortazavian, A. Bacterial nanocellulose: Biosynthesis and medical application. Biointerface Res. Appl. Chem. 2016, 6, 1511–1516. [Google Scholar]
  23. Berndt, S.; Wesarg, F.; Wiegand, C.; Kralisch, D.; Mueller, F.A. Antimicrobial porous hybrids consisting of bacterial nanocellulose and silver nanoparticles. Cellulose 2013, 20, 771–783. [Google Scholar] [CrossRef]
  24. Liyaskina, E.; Revin, V.; Paramonova, E.; Nazarkina, M.; Pestov, N.; Revina, N.; Kolesnikova, S. Nanomaterials from bacterial cellulose for antimicrobial wound dressing. J. Phys. Conf. Ser. 2017, 784, 012034. [Google Scholar] [CrossRef] [Green Version]
  25. Brown, A.J. XLIII—On an acetic ferment which forms cellulose. J. Chem. Soc. Trans. 1886, 49, 432–439. [Google Scholar] [CrossRef] [Green Version]
  26. Bacakova, L.; Pajorova, J.; Bacakova, M.; Skogberg, A.; Kallio, P.; Kolarova, K.; Svorcik, V. Versatile Application of Nanocellulose: From Industry to Skin Tissue Engineering and Wound Healing. Nanomaterials 2019, 9, 164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Martínez Ávila, H.; Schwarz, S.; Feldmann, E.-M.; Mantas, A.; von Bomhard, A.; Gatenholm, P.; Rotter, N. Biocompatibility evaluation of densified bacterial nanocellulose hydrogel as an implant material for auricular cartilage regeneration. Appl. Microbiol. Biotechnol. 2014, 98, 7423–7435. [Google Scholar] [CrossRef]
  28. Jozala, A.F.; de Lencastre-Novaes, L.C.; Lopes, A.M.; de Carvalho Santos-Ebinuma, V.; Mazzola, P.G.; Pessoa-Jr, A.; Grotto, D.; Gerenutti, M.; Chaud, M.V. Bacterial nanocellulose production and application: A 10-year overview. Appl. Microbiol. Biotechnol. 2016, 100, 2063–2072. [Google Scholar] [CrossRef] [Green Version]
  29. Habibi, Y. Key advances in the chemical modification of nanocelluloses. Chem. Soc. Rev. 2014, 43, 1519–1542. [Google Scholar] [CrossRef]
  30. Helenius, G.; Bäckdahl, H.; Bodin, A.; Nannmark, U.; Gatenholm, P.; Risberg, B. In vivo biocompatibility of bacterial cellulose. J. Biomed. Mater. Res. Part A 2006, 76A, 431–438. [Google Scholar] [CrossRef]
  31. Klemm, D.; Schumann, D.; Kramer, F.; Hessler, N.; Hornung, M.; Schmauder, H.-P.; Marsch, S. Nanocelluloses as innovative polymers in research and application. In Polysaccharides Ii; Klemm, D., Ed.; Springer: Berlin/Heidelberg, Germany, 2006; Volume 205, pp. 49–96. ISBN 978-3-540-37102-1. [Google Scholar]
  32. Siro, I.; Plackett, D. Microfibrillated cellulose and new nanocomposite materials: A review. Cellulose 2010, 17, 459–494. [Google Scholar] [CrossRef]
  33. Dufresne, A. Nanocellulose: From Nature to High Performance Tailored Materials, 2nd ed.; De Gruyter: Berlin, Germany, 2017. [Google Scholar]
  34. Hossain, A.B.M.S.; Uddin, M.; Nettoor Veettil, V.; Fawzi, M. Nano-cellulose based nano-coating biomaterial dataset using corn leaf biomass: An innovative biodegradable plant Biomaterial. Data Brief 2018, 17, 162–168. [Google Scholar] [CrossRef]
  35. Rashad, A.; Mohamed Ahmed, S.; Ojansivu, M.; Berstad, K.; Yassin, M.; Kivijärvi, T.; Heggset, E.B.; Syverud, K.; Mustafa, K. Coating 3D Printed Polycaprolactone Scaffolds with Nanocellulose Promotes Growth and Differentiation of Mesenchymal Stem Cells. Biomacromolecules 2018, 19, 4307–4319. [Google Scholar] [CrossRef]
  36. Korhonen, J.T.; Kettunen, M.; Ras, R.H.A.; Ikkala, O. Hydrophobic Nanocellulose Aerogels as Floating, Sustainable, Reusable, and Recyclable Oil Absorbents. ACS Appl. Mater. Interfaces 2011, 3, 1813–1816. [Google Scholar] [CrossRef]
  37. Wicklein, B.; Kocjan, A.; Salazar-Alvarez, G.; Carosio, F.; Camino, G.; Antonietti, M.; Bergstrom, L. Thermally insulating and fire-retardant lightweight anisotropic foams based on nanocellulose and graphene oxide. Nat. Nanotechnol. 2015, 10, 277–283. [Google Scholar] [CrossRef]
  38. De France, K.J.; Hoare, T.; Cranston, E.D. Review of Hydrogels and Aerogels Containing Nanocellulose. Chem. Mater. 2017, 29, 4609–4631. [Google Scholar] [CrossRef]
  39. Kim, J.-H.; Shim, B.S.; Kim, H.S.; Lee, Y.-J.; Min, S.-K.; Jang, D.; Abas, Z.; Kim, J. Review of nanocellulose for sustainable future materials. Int. J. Precis. Eng. Manuf.-Green Technol. 2015, 2, 197–213. [Google Scholar] [CrossRef] [Green Version]
  40. Auad, M.L.; Contos, V.S.; Nutt, S.; Aranguren, M.I.; Marcovich, N.E. Characterization of nanocellulose- reinforced shape memory polyurethanes. Polym. Int. 2008, 57, 651–659. [Google Scholar] [CrossRef]
  41. Czaja, W.K.; Young, D.J.; Kawecki, M.; Brown, R.M. The Future Prospects of Microbial Cellulose in Biomedical Applications. Biomacromolecules 2007, 8, 1–12. [Google Scholar] [CrossRef]
  42. Kamel, S. Pharmaceutical significance of cellulose: A review. Express Polym. Lett. 2008, 2, 758–778. [Google Scholar] [CrossRef]
  43. Ullah, H.; Santos, H.A.; Khan, T. Applications of bacterial cellulose in food, cosmetics and drug delivery. Cellulose 2016, 23, 2291–2314. [Google Scholar] [CrossRef]
  44. Mozafari, M.R. (Ed.) Nanocarrier Technologies: Frontiers of Nanotherapy; Springer Netherlands: Dordrecht, The Netherlands, 2006; ISBN 978-1-4020-5040-4. [Google Scholar]
  45. Pachuau, L. Application of Nanocellulose for Controlled Drug Delivery. In Nanocellulose and Nanohydrogel Matrices; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2017; pp. 1–19. ISBN 978-3-527-80383-5. [Google Scholar]
  46. Moscovici, M.; Hlevca, C.; Casarica, A.; Pavaloiu, R.-D. Nanocellulose and Nanogels as Modern Drug Delivery Systems. In Nanocellulose and Nanohydrogel Matrices; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2017; pp. 209–269. ISBN 978-3-527-80383-5. [Google Scholar]
  47. Andresen, M.; Stenstad, P.; Møretrø, T.; Langsrud, S.; Syverud, K.; Johansson, L.-S.; Stenius, P. Nonleaching Antimicrobial Films Prepared from Surface-Modified Microfibrillated Cellulose. Biomacromolecules 2007, 8, 2149–2155. [Google Scholar] [CrossRef]
  48. Takahashi, H.; Caputo, G.A.; Vemparala, S.; Kuroda, K. Synthetic Random Copolymers as a Molecular Platform To Mimic Host-Defense Antimicrobial Peptides. Bioconjug. Chem. 2017, 28, 1340–1350. [Google Scholar] [CrossRef]
  49. Muñoz-Bonilla, A.; Fernández-García, M. Polymeric materials with antimicrobial activity. Prog. Polym. Sci. 2012, 37, 281–339. [Google Scholar] [CrossRef]
  50. Lin, N.; Tang, J.; Dufresne, A.; Tam, M.K.C. (Eds.) Advanced Functional Materials from Nanopolysaccharides; Springer Series in Biomaterials Science and Engineering; Springer Singapore: Singapore, 2019; ISBN 9789811509124. [Google Scholar]
  51. Sun, B.; Hou, Q.; Liu, Z.; Ni, Y. Sodium periodate oxidation of cellulose nanocrystal and its application as a paper wet strength additive. Cellulose 2015, 22, 1135–1146. [Google Scholar] [CrossRef]
  52. Salam, A.; Lucia, L.A.; Jameel, H. Fluorine-based surface decorated cellulose nanocrystals as potential hydrophobic and oleophobic materials. Cellulose 2015, 22, 397–406. [Google Scholar] [CrossRef]
  53. Huang, J.-L.; Li, C.-J.; Gray, D.G. Functionalization of cellulose nanocrystal films via “thiol–ene” click reaction. RSC Adv. 2014, 4, 6965–6969. [Google Scholar] [CrossRef]
  54. Hemraz, U.D.; Campbell, K.A.; Burdick, J.S.; Ckless, K.; Boluk, Y.; Sunasee, R. Cationic Poly(2-aminoethylmethacrylate) and Poly(N-(2-aminoethylmethacrylamide) Modified Cellulose Nanocrystals: Synthesis, Characterization, and Cytotoxicity. Biomacromolecules 2015, 16, 319–325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Jorfi, M.; Foster, E.J. Recent advances in nanocellulose for biomedical applications. J. Appl. Polym. Sci. 2015, 132. [Google Scholar] [CrossRef]
  56. Čolić, M.; Tomić, S.; Bekić, M. Immunological aspects of nanocellulose. Immunol. Lett. 2020, 222, 80–89. [Google Scholar] [CrossRef]
  57. Čolić, M.; Mihajlović, D.; Mathew, A.; Naseri, N.; Kokol, V. Cytocompatibility and immunomodulatory properties of wood based nanofibrillated cellulose. Cellulose 2015, 22, 763–778. [Google Scholar] [CrossRef]
  58. Tomić, S.; Kokol, V.; Mihajlović, D.; Mirčić, A.; Čolić, M. Native cellulose nanofibrills induce immune tolerance in vitro by acting on dendritic cells. Sci. Rep. 2016, 6, 31618. [Google Scholar] [CrossRef]
  59. Tomić, S.; Ilić, N.; Kokol, V.; Gruden-Movsesijan, A.; Mihajlović, D.; Bekić, M.; Sofronić-Milosavljević, L.; Čolić, M.; Vučević, D. Functionalization-dependent effects of cellulose nanofibrils on tolerogenic mechanisms of human dendritic cells. Int. J. Nanomed. 2018, 13, 6941–6960. [Google Scholar] [CrossRef] [Green Version]
  60. Namvar, F.; Jawaid, M.; Tanir, P.M.; Mohamad, R.; Azizi, S.; Khodavandi, A.; Rahman, H.S.; Nayeri, M.D. Potential Use of Plant Fibres and their Composites for Biomedical Applications. BioResources 2014, 9, 5688–5706. [Google Scholar] [CrossRef]
  61. Pickering, K.L.; Efendy, M.G.A.; Le, T.M. A review of recent developments in natural fibre composites and their mechanical performance. Compos. Part A Appl. Sci. Manuf. 2016, 83, 98–112. [Google Scholar] [CrossRef] [Green Version]
  62. Cheung, H.; Ho, M.; Lau, K.; Cardona, F.; Hui, D. Natural fibre-reinforced composites for bioengineering and environmental engineering applications. Compos. Part B Eng. 2009, 40, 655–663. [Google Scholar] [CrossRef]
  63. Jia, B.; Li, Y.; Yang, B.; Xiao, D.; Zhang, S.; Rajulu, A.V.; Kondo, T.; Zhang, L.; Zhou, J. Effect of microcrystal cellulose and cellulose whisker on biocompatibility of cellulose-based electrospun scaffolds. Cellulose 2013, 20, 1911–1923. [Google Scholar] [CrossRef]
  64. Shi, Z.; Phillips, G.O.; Yang, G. Nanocellulose electroconductive composites. Nanoscale 2013, 5, 3194–3201. [Google Scholar] [CrossRef]
  65. Li, J.; Cha, R.; Mou, K.; Zhao, X.; Long, K.; Luo, H.; Zhou, F.; Jiang, X. Nanocellulose-Based Antibacterial Materials. Adv. Healthc. Mater. 2018, 7, 1800334. [Google Scholar] [CrossRef]
  66. Maneerung, T.; Tokura, S.; Rujiravanit, R. Impregnation of silver nanoparticles into bacterial cellulose for antimicrobial wound dressing. Carbohydr. Polym. 2008, 72, 43–51. [Google Scholar] [CrossRef]
  67. Luan, J.; Wu, J.; Zheng, Y.; Song, W.; Wang, G.; Guo, J.; Ding, X. Impregnation of silver sulfadiazine into bacterial cellulose for antimicrobial and biocompatible wound dressing. Biomed. Mater. 2012, 7, 065006. [Google Scholar] [CrossRef]
  68. Liu, C.; Yang, D.; Wang, Y.; Shi, J.; Jiang, Z. Fabrication of antimicrobial bacterial cellulose–Ag/AgCl nanocomposite using bacteria as versatile biofactory. J. Nanopart. Res. 2012, 14, 1084. [Google Scholar] [CrossRef]
  69. Xiong, R.; Lu, C.; Zhang, W.; Zhou, Z.; Zhang, X. Facile synthesis of tunable silver nanostructures for antibacterial application using cellulose nanocrystals. Carbohydr. Polym. 2013, 95, 214–219. [Google Scholar] [CrossRef]
  70. Fortunati, E.; Rinaldi, S.; Peltzer, M.; Bloise, N.; Visai, L.; Armentano, I.; Jiménez, A.; Latterini, L.; Kenny, J.M. Nano-biocomposite films with modified cellulose nanocrystals and synthesized silver nanoparticles. Carbohydr. Polym. 2014, 101, 1122–1133. [Google Scholar] [CrossRef] [Green Version]
  71. Gan, I.; Chow, W.S. Antimicrobial poly(lactic acid)/cellulose bionanocomposite for food packaging application: A review. Food Packaging and Shelf Life 2018, 17, 150–161. [Google Scholar] [CrossRef]
  72. Jipa, I.M.; Stoica-Guzun, A.; Stroescu, M. Controlled release of sorbic acid from bacterial cellulose based mono and multilayer antimicrobial films. LWT 2012, 47, 400–406. [Google Scholar] [CrossRef]
  73. Fernandes, S.C.M.; Sadocco, P.; Alonso-Varona, A.; Palomares, T.; Eceiza, A.; Silvestre, A.J.D.; Mondragon, I.; Freire, C.S.R. Bioinspired Antimicrobial and Biocompatible Bacterial Cellulose Membranes Obtained by Surface Functionalization with Aminoalkyl Groups. ACS Appl. Mater. Interfaces 2013, 5, 3290–3297. [Google Scholar] [CrossRef]
  74. Butchosa, N.; Brown, C.; Larsson, P.T.; Berglund, L.A.; Bulone, V.; Zhou, Q. Nanocomposites of bacterial cellulose nanofibers and chitin nanocrystals: Fabrication, characterization and bactericidal activity. Green Chem. 2013, 15, 3404–3413. [Google Scholar] [CrossRef]
  75. Zhang, T.; Zhou, P.; Zhan, Y.; Shi, X.; Lin, J.; Du, Y.; Li, X.; Deng, H. Pectin/lysozyme bilayers layer-by-layer deposited cellulose nanofibrous mats for antibacterial application. Carbohydr. Polym. 2015, 117, 687–693. [Google Scholar] [CrossRef]
  76. Shao, W.; Liu, H.; Liu, X.; Wang, S.; Zhang, R. Anti-bacterial performances and biocompatibility of bacterial cellulose/graphene oxide composites. RSC Adv. 2014, 5, 4795–4803. [Google Scholar] [CrossRef]
  77. Sun, X.; Zhang, L.; Cao, Z.; Deng, Y.; Liu, L.; Fong, H.; Sun, Y. Electrospun composite nanofiber fabrics containing uniformly dispersed antimicrobial agents as an innovative type of polymeric materials with superior antimicrobial efficacy. ACS Appl. Mater. Interfaces 2010, 2, 952–956. [Google Scholar] [CrossRef]
  78. Gomez-Orellana, I. Strategies to improve oral drug bioavailability. Expert Opin. Drug Deliv. 2005, 2, 419–433. [Google Scholar] [CrossRef]
  79. Boddupalli, B.M.; Mohammed, Z.N.K.; Nath, R.A.; Banji, D. Mucoadhesive drug delivery system: An overview. J. Adv. Pharm. Technol. Res. 2010, 1, 381–387. [Google Scholar] [CrossRef] [Green Version]
  80. Smart, J.D. The basics and underlying mechanisms of mucoadhesion. Adv. Drug Deliv. Rev. 2005, 57, 1556–1568. [Google Scholar] [CrossRef]
  81. Shaikh, R.; Raj Singh, T.R.; Garland, M.J.; Woolfson, A.D.; Donnelly, R.F. Mucoadhesive drug delivery systems. J. Pharm. Bioallied Sci. 2011, 3, 89–100. [Google Scholar] [CrossRef] [PubMed]
  82. Ibrahim, M.; Verma, R.; Garcia-Contreras, L. Inhalation drug delivery devices: Technology update. Med. Devices 2015, 8, 131–139. [Google Scholar] [CrossRef] [Green Version]
  83. Peterfreund, R.A.; Philip, J.H. Critical parameters in drug delivery by intravenous infusion. Expert Opin. Drug Deliv. 2013, 10, 1095–1108. [Google Scholar] [CrossRef] [PubMed]
  84. Prausnitz, M.R.; Langer, R. Transdermal drug delivery. Nat. Biotechnol. 2008, 26, 1261–1268. [Google Scholar] [CrossRef] [PubMed]
  85. Van Smeden, J.; Janssens, M.; Gooris, G.S.; Bouwstra, J.A. The important role of stratum corneum lipids for the cutaneous barrier function. Biochim. Biophys. Acta 2014, 1841, 295–313. [Google Scholar] [CrossRef]
  86. Maher, S.; Brayden, D.J.; Casettari, L.; Illum, L. Application of Permeation Enhancers in Oral Delivery of Macromolecules: An Update. Pharmaceutics 2019, 11, 41. [Google Scholar] [CrossRef] [Green Version]
  87. Lane, M.E. Skin penetration enhancers. Int. J. Pharm. 2013, 447, 12–21. [Google Scholar] [CrossRef]
  88. Tran, M.; Wang, C. Semi-solid materials for controlled release drug formulation: Current status and future prospects. Front. Chem. Sci. Eng. 2014, 8, 225–232. [Google Scholar] [CrossRef]
  89. Ioelovich, M. Chapter 9 - Nanocellulose—Fabrication, structure, properties, and application in the area of care and cure. In Fabrication and Self-Assembly of Nanobiomaterials; Grumezescu, A.M., Ed.; William Andrew Publishing: Norwich, NY, USA, 2016; pp. 243–288. ISBN 978-0-323-41533-0. [Google Scholar]
  90. Börjesson, M.; Westman, G. Crystalline Nanocellulose—Preparation, Modification, and Properties. Cellulose: Fundamental Aspects and Current Trends; IntechOpen: London, UK, 2015. [Google Scholar] [CrossRef] [Green Version]
  91. Chang, C.; Hou, J.; Chang, P.R.; Huang, J. Structure and Properties of Cellulose Nanocrystals. In Nanocellulose: From Fundamentals to Advanced Materials; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2019; pp. 21–52. ISBN 978-3-527-80743-7. [Google Scholar]
  92. Daud, J.B.; Lee, K.-Y. Surface Modification of Nanocellulose. In Handbook of Nanocellulose and Cellulose Nanocomposites; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2017; pp. 101–122. ISBN 978-3-527-68997-2. [Google Scholar]
  93. Islam, M.T.; Alam, M.M.; Zoccola, M. Review on modification of nanocellulose for application in composites. Int. J. Inov. Res. Sci. Eng. Technol. 2013, 2, 5444–5451. [Google Scholar]
  94. Tan, T.H.; Lee, H.V.; Yehya Dabdawb, W.A.; Hamid, S.B.B.O.A.A. Chapter 5 - A review of nanocellulose in the drug-delivery system. In Materials for Biomedical Engineering; Holban, A.-M., Grumezescu, A.M., Eds.; Elsevier: Amsterdam, The Netherlands, 2019; pp. 131–164. ISBN 978-0-12-816913-1. [Google Scholar]
  95. Kolakovic, R.; Peltonen, L.; Laukkanen, A.; Hirvonen, J.; Laaksonen, T. Nanofibrillar cellulose films for controlled drug delivery. Eur. J. Pharm. Biopharm. 2012, 82, 308–315. [Google Scholar] [CrossRef]
  96. Salimi, S.; Sotudeh-Gharebagh, R.; Zarghami, R.; Chan, S.Y.; Yuen, K.H. Production of Nanocellulose and Its Applications in Drug Delivery: A Critical Review. ACS Sustain. Chem. Eng. 2019, 7, 15800–15827. [Google Scholar] [CrossRef]
  97. Plackett, D.V.; Letchford, K.; Jackson, J.K.; Burt, H.M. A review of nanocellulose as a novel vehicle for drug delivery. Nord. Pulp Paper Res. J. 2014, 29, 105–118. [Google Scholar] [CrossRef]
  98. Kiliona, K.P.S.; Lwal, A.L.J.; Tao, H.; Lin, N. Surface Modification with Grafting Functional Molecules on Nanopolysaccharides. In Advanced Functional Materials from Nanopolysaccharides; Lin, N., Tang, J., Dufresne, A., Tam, M.K.C., Eds.; Springer Series in Biomaterials Science and Engineering; Springer: Singapore, 2019; pp. 55–85. ISBN 9789811509131. [Google Scholar]
  99. Lin, N.; Gèze, A.; Wouessidjewe, D.; Huang, J.; Dufresne, A. Biocompatible Double-Membrane Hydrogels from Cationic Cellulose Nanocrystals and Anionic Alginate as Complexing Drugs Codelivery. ACS Appl. Mater. Interfaces 2016, 8, 6880–6889. [Google Scholar] [CrossRef]
  100. Ndong Ntoutoume, G.M.A.; Granet, R.; Mbakidi, J.P.; Brégier, F.; Léger, D.Y.; Fidanzi-Dugas, C.; Lequart, V.; Joly, N.; Liagre, B.; Chaleix, V.; et al. Development of curcumin–cyclodextrin/cellulose nanocrystals complexes: New anticancer drug delivery systems. Bioorganic Med. Chem. Lett. 2016, 26, 941–945. [Google Scholar] [CrossRef]
  101. You, J.; Cao, J.; Zhao, Y.; Zhang, L.; Zhou, J.; Chen, Y. Improved Mechanical Properties and Sustained Release Behavior of Cationic Cellulose Nanocrystals Reinforeced Cationic Cellulose Injectable Hydrogels. Biomacromolecules 2016, 17, 2839–2848. [Google Scholar] [CrossRef] [PubMed]
  102. Golshan, M.; Salami-Kalajahi, M.; Roghani-Mamaqani, H.; Mohammadi, M. Poly(propylene imine) dendrimer-grafted nanocrystalline cellulose: Doxorubicin loading and release behavior. Polymer 2017, 117, 287–294. [Google Scholar] [CrossRef] [Green Version]
  103. Gorgieva, S.; Vivod, V.; Maver, U.; Gradišnik, L.; Dolenšek, J.; Kokol, V. Internalization of (bis)phosphonate-modified cellulose nanocrystals by human osteoblast cells. Cellulose 2017, 24, 4235–4252. [Google Scholar] [CrossRef] [Green Version]
  104. Supramaniam, J.; Adnan, R.; Mohd Kaus, N.H.; Bushra, R. Magnetic nanocellulose alginate hydrogel beads as potential drug delivery system. Int. J. Biol. Macromol. 2018, 118, 640–648. [Google Scholar] [CrossRef] [PubMed]
  105. Tummala, G.K. Hydrogels of Poly(vinyl alcohol) and Nanocellulose for Ophthalmic Applications: Synthesis, Characterization, Biocompatibility and Drug Delivery Studies. Ph.D. Thesis, Acta Universitatis Upsaliensis, Uppsala, Sweden, 2018. [Google Scholar]
  106. Md Abu, T.; Zahan, K.A.; Rajaie, M.A.; Leong, C.R.; Ab Rashid, S.; Mohd Nor Hamin, N.S.; Tan, W.N.; Tong, W.Y. Nanocellulose as drug delivery system for honey as antimicrobial wound dressing. Mater. Today Proc. 2020, 31, 14–17. [Google Scholar] [CrossRef]
  107. Zainuddin, N.; Ahmad, I.; Kargarzadeh, H.; Ramli, S. Hydrophobic kenaf nanocrystalline cellulose for the binding of curcumin. Carbohydr. Polym. 2017, 163, 261–269. [Google Scholar] [CrossRef]
  108. Tong, W.Y.; bin Abdullah, A.Y.K.; binti Rozman, N.A.S.; bin Wahid, M.I.A.; Hossain, M.S.; Ring, L.C.; Lazim, Y.; Tan, W.-N. Antimicrobial wound dressing film utilizing cellulose nanocrystal as drug delivery system for curcumin. Cellulose 2018, 25, 631–638. [Google Scholar] [CrossRef]
  109. Udeni Gunathilake, T.M.S.; Ching, Y.C.; Chuah, C.H. Enhancement of Curcumin Bioavailability Using Nanocellulose Reinforced Chitosan Hydrogel. Polymers 2017, 9, 64. [Google Scholar] [CrossRef] [Green Version]
  110. Sampath Udeni Gunathilake, T.M.; Ching, Y.C.; Chuah, C.H.; Illias, H.A.; Ching, K.Y.; Singh, R.; Nai-Shang, L. Influence of a nonionic surfactant on curcumin delivery of nanocellulose reinforced chitosan hydrogel. Int. J. Biol. Macromol. 2018, 118, 1055–1064. [Google Scholar] [CrossRef] [PubMed]
  111. Ching, Y.C.; Gunathilake, T.M.S.U.; Chuah, C.H.; Ching, K.Y.; Singh, R.; Liou, N.-S. Curcumin/Tween 20-incorporated cellulose nanoparticles with enhanced curcumin solubility for nano-drug delivery: Characterization and in vitro evaluation. Cellulose 2019, 26, 5467–5481. [Google Scholar] [CrossRef]
  112. Li, N.; Zhang, H.; Xiao, Y.; Huang, Y.; Xu, M.; You, D.; Lu, W.; Yu, J. Fabrication of Cellulose-Nanocrystal-Based Folate Targeted Nanomedicine via Layer-by-Layer Assembly with Lysosomal pH-Controlled Drug Release into the Nucleus. Biomacromolecules 2019, 20, 937–948. [Google Scholar] [CrossRef] [PubMed]
  113. Hivechi, A.; Bahrami, S.H.; Siegel, R.A. Drug release and biodegradability of electrospun cellulose nanocrystal reinforced polycaprolactone. Mater. Sci. Eng. C 2019, 94, 929–937. [Google Scholar] [CrossRef]
  114. Löbmann, K.; Svagan, A.J. Cellulose nanofibers as excipient for the delivery of poorly soluble drugs. Int. J. Pharm. 2017, 533, 285–297. [Google Scholar] [CrossRef]
  115. Svagan, A.J.; Benjamins, J.-W.; Al-Ansari, Z.; Shalom, D.B.; Müllertz, A.; Wågberg, L.; Löbmann, K. Solid cellulose nanofiber based foams—Towards facile design of sustained drug delivery systems. J. Control. Release 2016, 244, 74–82. [Google Scholar] [CrossRef]
  116. Löbmann, K.; Wohlert, J.; Müllertz, A.; Wågberg, L.; Svagan, A.J. Cellulose Nanopaper and Nanofoam for Patient-Tailored Drug Delivery. Adv. Mater. Interfaces 2017, 4, 1600655. [Google Scholar] [CrossRef]
  117. Bhandari, J.; Mishra, H.; Mishra, P.K.; Wimmer, R.; Ahmad, F.J.; Talegaonkar, S. Cellulose nanofiber aerogel as a promising biomaterial for customized oral drug delivery. Int. J. Nanomed. 2017, 12, 2021–2031. [Google Scholar] [CrossRef] [Green Version]
  118. Paukkonen, H.; Ukkonen, A.; Szilvay, G.; Yliperttula, M.; Laaksonen, T. Hydrophobin-nanofibrillated cellulose stabilized emulsions for encapsulation and release of BCS class II drugs. Eur. J. Pharm. Sci. 2017, 100, 238–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Svagan, A.J.; Müllertz, A.; Löbmann, K. Floating solid cellulose nanofibre nanofoams for sustained release of the poorly soluble model drug furosemide. J. Pharm. Pharmacol. 2017, 69, 1477–1484. [Google Scholar] [CrossRef] [PubMed]
  120. Fakhri, A.; Tahami, S.; Nejad, P.A. Preparation and characterization of Fe3O4-Ag2O quantum dots decorated cellulose nanofibers as a carrier of anticancer drugs for skin cancer. J. Photochem. Photobiol. B Biol. 2017, 175, 83–88. [Google Scholar] [CrossRef] [PubMed]
  121. Paukkonen, H.; Kunnari, M.; Laurén, P.; Hakkarainen, T.; Auvinen, V.-V.; Oksanen, T.; Koivuniemi, R.; Yliperttula, M.; Laaksonen, T. Nanofibrillar cellulose hydrogels and reconstructed hydrogels as matrices for controlled drug release. Int. J. Pharm. 2017, 532, 269–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Guo, T.; Pei, Y.; Tang, K.; He, X.; Huang, J.; Wang, F. Mechanical and drug release properties of alginate beads reinforced with cellulose. J. Appl. Polym. Sci. 2017, 134. [Google Scholar] [CrossRef]
  123. Poonguzhali, R.; Khaleel Basha, S.; Sugantha Kumari, V. Synthesis of alginate/nanocellulose bionanocomposite for in vitro delivery of ampicillin. Polym. Bull. 2018, 75, 4165–4173. [Google Scholar] [CrossRef] [Green Version]
  124. Liu, Y.; Sui, Y.; Liu, C.; Liu, C.; Wu, M.; Li, B.; Li, Y. A physically crosslinked polydopamine/nanocellulose hydrogel as potential versatile vehicles for drug delivery and wound healing. Carbohydr. Polym. 2018, 188, 27–36. [Google Scholar] [CrossRef]
  125. Sarkar, G.; Orasugh, J.T.; Saha, N.R.; Roy, I.; Bhattacharyya, A.; Chattopadhyay, A.K.; Rana, D.; Chattopadhyay, D. Cellulose nanofibrils/chitosan based transdermal drug delivery vehicle for controlled release of ketorolac tromethamine. New J. Chem. 2017, 41, 15312–15319. [Google Scholar] [CrossRef]
  126. Orasugh, J.T.; Saha, N.R.; Rana, D.; Sarkar, G.; Mollick, M.M.R.; Chattoapadhyay, A.; Mitra, B.C.; Mondal, D.; Ghosh, S.K.; Chattopadhyay, D. Jute cellulose nano-fibrils/hydroxypropylmethylcellulose nanocomposite: A novel material with potential for application in packaging and transdermal drug delivery system. Ind. Crops Prod. 2018, 112, 633–643. [Google Scholar] [CrossRef]
  127. Auvinen, V.-V.; Virtanen, J.; Merivaara, A.; Virtanen, V.; Laurén, P.; Tuukkanen, S.; Laaksonen, T. Modulating sustained drug release from nanocellulose hydrogel by adjusting the inner geometry of implantable capsules. J. Drug Deliv. Sci. Technol. 2020, 57, 101625. [Google Scholar] [CrossRef]
  128. Gun’ko, V.M.; Savina, I.N.; Mikhalovsky, S.V. Cryogels: Morphological, structural and adsorption characterisation. Adv. Colloid Interface Sci. 2013, 187–188, 1–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Li, J.; Wang, Y.; Zhang, L.; Xu, Z.; Dai, H.; Wu, W. Nanocellulose/Gelatin Composite Cryogels for Controlled Drug Release. ACS Sustain. Chem. Eng. 2019, 7, 6381–6389. [Google Scholar] [CrossRef]
  130. Meneguin, A.B.; Ferreira Cury, B.S.; dos Santos, A.M.; Franco, D.F.; Barud, H.S.; da Silva Filho, E.C. Resistant starch/pectin free-standing films reinforced with nanocellulose intended for colonic methotrexate release. Carbohydr. Polym. 2017, 157, 1013–1023. [Google Scholar] [CrossRef] [PubMed]
  131. Fiorati, A.; Turco, G.; Travan, A.; Caneva, E.; Pastori, N.; Cametti, M.; Punta, C.; Melone, L. Mechanical and Drug Release Properties of Sponges from Cross-linked Cellulose Nanofibers. ChemPlusChem 2017, 82, 848–858. [Google Scholar] [CrossRef] [PubMed]
  132. Fiorati, A.; Contessi Negrini, N.; Baschenis, E.; Altomare, L.; Faré, S.; Giacometti Schieroni, A.; Piovani, D.; Mendichi, R.; Ferro, M.; Castiglione, F.; et al. TEMPO-Nanocellulose/Ca2+ Hydrogels: Ibuprofen Drug Diffusion and In Vitro Cytocompatibility. Materials 2020, 13, 183. [Google Scholar] [CrossRef] [Green Version]
  133. Alkhatib, Y.; Dewaldt, M.; Moritz, S.; Nitzsche, R.; Kralisch, D.; Fischer, D. Controlled extended octenidine release from a bacterial nanocellulose/Poloxamer hybrid system. Eur. J. Pharm. Biopharm. 2017, 112, 164–176. [Google Scholar] [CrossRef]
  134. Medhi, P.; Olatunji, O.; Nayak, A.; Uppuluri, C.T.; Olsson, R.T.; Nalluri, B.N.; Das, D.B. Lidocaine-loaded fish scale-nanocellulose biopolymer composite microneedles. AAPS PharmSciTech 2017, 18, 1488–1494. [Google Scholar] [CrossRef]
  135. Saïdi, L.; Vilela, C.; Oliveira, H.; Silvestre, A.J.D.; Freire, C.S.R. Poly(N-methacryloyl glycine)/nanocellulose composites as pH-sensitive systems for controlled release of diclofenac. Carbohydr. Polym. 2017, 169, 357–365. [Google Scholar] [CrossRef]
  136. Ratnayake, W.M.K.M.; Damunupola, J.W.; Rajapakse, S.; Jayasundera, A.C.A. Nanocellulose-Protein Matrices: A Model System for Controlled Drug Delivery. In Proceedings of the 5th International Conference on Nanoscience and Nanotechnology, Dubai, UAE, 16 October 2017; 2018; Volume 5, pp. 1–13. [Google Scholar]
  137. Fey, C.; Betz, J.; Rosenbaum, C.; Kralisch, D.; Vielreicher, M.; Friedrich, O.; Metzger, M.; Zdzieblo, D. Bacterial nanocellulose as novel carrier for intestinal epithelial cells in drug delivery studies. Mater. Sci. Eng. C 2020, 109, 110613. [Google Scholar] [CrossRef]
  138. Silva, N.H.C.S.; Mota, J.P.; Santos de Almeida, T.; Carvalho, J.P.F.; Silvestre, A.J.D.; Vilela, C.; Rosado, C.; Freire, C.S.R. Topical Drug Delivery Systems Based on Bacterial Nanocellulose: Accelerated Stability Testing. Int. J. Mol. Sci. 2020, 21, 1262. [Google Scholar] [CrossRef] [Green Version]
  139. Abba, M.; Ibrahim, Z.; Chong, C.S.; Zawawi, N.A.; Kadir, M.R.A.; Yusof, A.H.M.; Razak, S.I.A. Transdermal Delivery of Crocin Using Bacterial Nanocellulose Membrane. Fibers Polym. 2019, 20, 2025–2031. [Google Scholar] [CrossRef]
  140. Alavizadeh, S.H.; Hosseinzadeh, H. Bioactivity assessment and toxicity of crocin: A comprehensive review. Food Chem. Toxicol. 2014, 64, 65–80. [Google Scholar] [CrossRef] [PubMed]
  141. Roy, D.; Knapp, J.S.; Guthrie, J.T.; Perrier, S. Antibacterial cellulose fiber via RAFT surface graft polymerization. Biomacromolecules 2008, 9, 91–99. [Google Scholar] [CrossRef] [PubMed]
  142. Rai, M.; Yadav, A.; Gade, A. Silver nanoparticles as a new generation of antimicrobials. Biotechnol. Adv. 2009, 27, 76–83. [Google Scholar] [CrossRef] [PubMed]
  143. Gao, C.; Yan, T.; Du, J.; He, F.; Luo, H.; Wan, Y. Introduction of broad spectrum antibacterial properties to bacterial cellulose nanofibers via immobilising ε-polylysine nanocoatings. Food Hydrocoll. 2014, 36, 204–211. [Google Scholar] [CrossRef]
  144. Panchal, P.; Ogunsona, E.; Mekonnen, T. Trends in Advanced Functional Material Applications of Nanocellulose. Processes 2019, 7, 10. [Google Scholar] [CrossRef] [Green Version]
  145. Appendini, P.; Hotchkiss, J.H. Review of antimicrobial food packaging. Innov. Food Sci. Emerg. Technol. 2002, 3, 113–126. [Google Scholar] [CrossRef]
  146. Gudikandula, K.; Maringanti, S.C. Synthesis of silver nanoparticles by chemical and biological methods and their antimicrobial properties. J. Exp. Nanosci. 2016, 11, 714–721. [Google Scholar] [CrossRef]
  147. Panyala, N.R.; Pena, E.M.; Havel, J. Silver or silver nanoparticles: A hazardous threat to the environment and human health? J. Appl. Biomed. 2008, 6, 117–129. [Google Scholar] [CrossRef] [Green Version]
  148. Cho, K.-H.; Park, J.-E.; Osaka, T.; Park, S.-G. The study of antimicrobial activity and preservative effects of nanosilver ingredient. Electrochim. Acta 2005, 51, 956–960. [Google Scholar] [CrossRef]
  149. Morones, J.R.; Elechiguerra, J.L.; Camacho, A.; Holt, K.; Kouri, J.B.; Ramírez, J.T.; Yacaman, M.J. The bactericidal effect of silver nanoparticles. Nanotechnology 2005, 16, 2346–2353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Yoon, K.-Y.; Byeon, J.H.; Park, J.-H.; Ji, J.H.; Bae, G.N.; Hwang, J. Antimicrobial Characteristics of Silver Aerosol Nanoparticles against Bacillus subtilis Bioaerosols. Environ. Eng. Sci. 2008, 25, 289–294. [Google Scholar] [CrossRef] [Green Version]
  151. Shrivastava, S.; Bera, T.; Roy, A.; Singh, G.; Ramachandrarao, P.; Dash, D. Characterization of enhanced antibacterial effects of novel silver nanoparticles. Nanotechnology 2007, 18, 225103. [Google Scholar] [CrossRef]
  152. Panáček, A.; Kvítek, L.; Prucek, R.; Kolář, M.; Večeřová, R.; Pizúrová, N.; Sharma, V.K.; Nevěčná, T.; Zbořil, R. Silver Colloid Nanoparticles:  Synthesis, Characterization, and Their Antibacterial Activity. J. Phys. Chem. B 2006, 110, 16248–16253. [Google Scholar] [CrossRef]
  153. Sharma, V.K.; Yngard, R.A.; Lin, Y. Silver nanoparticles: Green synthesis and their antimicrobial activities. Adv. Colloid Interface Sci. 2009, 145, 83–96. [Google Scholar] [CrossRef]
  154. Barud, H.; Regiani, T.; Marques, R.; Lustri, W.; Messaddeq, Y.; Ribeiro, S. Antimicrobial Bacterial Cellulose-Silver Nanoparticles Composite Membranes. J. Nanomater. 2011, 2011, 721631. [Google Scholar] [CrossRef] [Green Version]
  155. Yang, G.; Xie, J.; Hong, F.; Cao, Z.; Yang, X. Antimicrobial activity of silver nanoparticle impregnated bacterial cellulose membrane: Effect of fermentation carbon sources of bacterial cellulose. Carbohydr. Polym. 2012, 87, 839–845. [Google Scholar] [CrossRef]
  156. Amini, E.; Azadfallah, M.; Layeghi, M.; Talaei-Hassanloui, R. Silver-nanoparticle-impregnated cellulose nanofiber coating for packaging paper. Cellulose 2016, 23, 557–570. [Google Scholar] [CrossRef]
  157. Mohite, B.V.; Patil, S.V. In situ development of nanosilver-impregnated bacterial cellulose for sustainable released antimicrobial wound dressing. J. Appl. Biomater. Funct. Mater. 2016, 14, e53–e58. [Google Scholar] [CrossRef] [Green Version]
  158. Sygnatowicz, M.; Keyshar, K.; Tiwari, A. Antimicrobial properties of silver-doped hydroxyapatite nano-powders and thin films. JOM 2010, 62, 65–70. [Google Scholar] [CrossRef]
  159. Joshy, M.I.A.; Elayaraja, K.; Sakthivel, N.; Chandra, V.S.; Shanthini, G.M.; Kalkura, S.N. Freeze dried cross linking free biodegradable composites with microstructures for tissue engineering and drug delivery application. Mater. Sci. Eng. C 2013, 33, 466–474. [Google Scholar] [CrossRef] [PubMed]
  160. Dubnika, A.; Loca, D.; Rudovica, V.; Parekh, M.B.; Berzina-Cimdina, L. Functionalized silver doped hydroxyapatite scaffolds for controlled simultaneous silver ion and drug delivery. Ceram. Int. 2017, 43, 3698–3705. [Google Scholar] [CrossRef]
  161. Yadollahi, M.; Farhoudian, S.; Namazi, H. One-pot synthesis of antibacterial chitosan/silver bio-nanocomposite hydrogel beads as drug delivery systems. Int. J. Biol. Macromol. 2015, 79, 37–43. [Google Scholar] [CrossRef] [PubMed]
  162. Kim, J.H.; Kim, S.I.; Kwon, I.B.; Kim, M.H.; Lim, J.I. Simple fabrication of silver hybridized porous chitosan-based patch for transdermal drug-delivery system. Mater. Lett. 2013, 95, 48–51. [Google Scholar] [CrossRef]
  163. Tran, C.; Prosenc, F.; Franko, M.; Benzi, G. One-Pot Synthesis of Biocompatible Silver Nanoparticle Composites from Cellulose and Keratin: Characterization and Antimicrobial Activity. ACS Appl. Mater. Interfaces 2016, 8, 34791–34801. [Google Scholar] [CrossRef] [Green Version]
  164. Dallas, P.; Sharma, V.K.; Zboril, R. Silver polymeric nanocomposites as advanced antimicrobial agents: Classification, synthetic paths, applications, and perspectives. Adv. Colloid Interface Sci. 2011, 166, 119–135. [Google Scholar] [CrossRef]
  165. Lizundia, E.; Goikuria, U.; Vilas, J.L.; Cristofaro, F.; Bruni, G.; Fortunati, E.; Armentano, I.; Visai, L.; Torre, L. Metal Nanoparticles Embedded in Cellulose Nanocrystal Based Films: Material Properties and Post-use Analysis. Biomacromolecules 2018, 19, 2618–2628. [Google Scholar] [CrossRef]
  166. Rasmussen, J.W.; Martinez, E.; Louka, P.; Wingett, D.G. Zinc oxide nanoparticles for selective destruction of tumor cells and potential for drug delivery applications. Expert Opin. Drug Deliv. 2010, 7, 1063–1077. [Google Scholar] [CrossRef] [Green Version]
  167. Yuan, Q.; Hein, S.; Misra, R.D.K. New generation of chitosan-encapsulated ZnO quantum dots loaded with drug: Synthesis, characterization and in vitro drug delivery response. Acta Biomater. 2010, 6, 2732–2739. [Google Scholar] [CrossRef]
  168. Barick, K.C.; Nigam, S.; Bahadur, D. Nanoscale assembly of mesoporous ZnO: A potential drug carrier. J. Mater. Chem. 2010, 20, 6446–6452. [Google Scholar] [CrossRef]
  169. Zhang, H.; Wang, C.; Chen, B.; Wang, X. Daunorubicin-TiO2 nanocomposites as a “smart” pH-responsive drug delivery system. Int. J. Nanomed. 2012, 7, 235–242. [Google Scholar] [CrossRef]
  170. Xiong, H.-M. ZnO Nanoparticles Applied to Bioimaging and Drug Delivery. Adv. Mater. 2013, 25, 5329–5335. [Google Scholar] [CrossRef] [PubMed]
  171. El-Wakil, N.A.; Hassan, E.A.; Abou-Zeid, R.E.; Dufresne, A. Development of wheat gluten/nanocellulose/titanium dioxide nanocomposites for active food packaging. Carbohydr. Polym. 2015, 124, 337–346. [Google Scholar] [CrossRef] [PubMed]
  172. Galkina, O.L.; Önneby, K.; Huang, P.; Ivanov, V.K.; Agafonov, A.V.; Seisenbaeva, G.A.; Kessler, V.G. Antibacterial and photochemical properties of cellulose nanofiber–titania nanocomposites loaded with two different types of antibiotic medicines. J. Mater. Chem. B 2015, 3, 7125–7134. [Google Scholar] [CrossRef] [PubMed]
  173. Barua, S.; Das, G.; Aidew, L.; Buragohain, A.K.; Karak, N. Copper–copper oxide coated nanofibrillar cellulose: A promising biomaterial. RSC Adv. 2013, 3, 14997–15004. [Google Scholar] [CrossRef]
  174. Lefatshe, K.; Muiva, C.M.; Kebaabetswe, L.P. Extraction of nanocellulose and in-situ casting of ZnO/cellulose nanocomposite with enhanced photocatalytic and antibacterial activity. Carbohydr. Polym. 2017, 164, 301–308. [Google Scholar] [CrossRef]
  175. Martins, N.C.T.; Freire, C.S.R.; Neto, C.P.; Silvestre, A.J.D.; Causio, J.; Baldi, G.; Sadocco, P.; Trindade, T. Antibacterial paper based on composite coatings of nanofibrillated cellulose and ZnO. Colloids Surf. A Physicochem. Eng. Asp. 2013, 417, 111–119. [Google Scholar] [CrossRef]
  176. Ul-Islam, M.; Khattak, W.A.; Ullah, M.W.; Khan, S.; Park, J.K. Synthesis of regenerated bacterial cellulose-zinc oxide nanocomposite films for biomedical applications. Cellulose 2014, 21, 433–447. [Google Scholar] [CrossRef]
  177. Katepetch, C.; Rujiravanit, R.; Tamura, H. Formation of nanocrystalline ZnO particles into bacterial cellulose pellicle by ultrasonic-assisted in situ synthesis. Cellulose 2013, 20, 1275–1292. [Google Scholar] [CrossRef]
  178. Mirtalebi, S.S.; Almasi, H.; Alizadeh Khaledabad, M. Physical, morphological, antimicrobial and release properties of novel MgO-bacterial cellulose nanohybrids prepared by in-situ and ex-situ methods. Int. J. Biol. Macromol. 2019, 128, 848–857. [Google Scholar] [CrossRef]
  179. Wang, J.; Vermerris, W. Antimicrobial Nanomaterials Derived from Natural Products—A Review. Materials 2016, 9, 255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Vilela, C.; Moreirinha, C.; Domingues, E.M.; Figueiredo, F.M.L.; Almeida, A.; Freire, C.S.R. Antimicrobial and Conductive Nanocellulose-Based Films for Active and Intelligent Food Packaging. Nanomaterials 2019, 9, 980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Sun, X.; Wu, Q.; Zhang, X.; Ren, S.; Lei, T.; Li, W.; Xu, G.; Zhang, Q. Nanocellulose films with combined cellulose nanofibers and nanocrystals: Tailored thermal, optical and mechanical properties. Cellulose 2018, 25, 1103–1115. [Google Scholar] [CrossRef]
  182. Halib, N.; Ahmad, I. Nanocellulose: Insight into health and medical applications. In Handbook of Ecomaterials; Springer International Publishing: Cham, Switzerland, 2019; pp. 1345–1363. [Google Scholar] [CrossRef]
  183. Bielecki, S.; Kalinowska, H.; Krystynowicz, A.; Kubiak, K.; Kołodziejczyk, M.; De Groeve, M. Wound dressings and cosmetic materials from bacterial nanocellulose. In Bacterial NanoCellulose: A Sophisticated Multifunctional Material; CRC Press: Boca Raton, FL, USA, 2016; pp. 157–174. [Google Scholar]
  184. Ioelovich, M.; Figovsky, O. Nano-cellulose as Promising Biocarrier. J. Mater. Res. 2008, 47–50, 1286–1289. [Google Scholar] [CrossRef]
  185. Ludwicka, K.; Jedrzejczak-Krzepkowska, M.; Kubiak, K.; Kolodziejczyk, M.; Pankiewicz, T.; Bielecki, S. Chapter 9 - Medical and Cosmetic Applications of Bacterial NanoCellulose. In Bacterial Nanocellulose; Gama, M., Dourado, F., Bielecki, S., Eds.; Elsevier: Amsterdam, The Netherlands, 2016; pp. 145–165. ISBN 978-0-444-63458-0. [Google Scholar]
  186. Dumanli, A.G. Nanocellulose and its Composites for Biomedical Applications. Curr. Med. Chem. 2017, 24, 512–528. [Google Scholar] [CrossRef] [PubMed]
  187. Wei, B.; Yang, G.; Hong, F. Preparation and evaluation of a kind of bacterial cellulose dry films with antibacterial properties. Carbohydr. Polym. 2011, 84, 533–538. [Google Scholar] [CrossRef]
  188. Wiegand, C.; Moritz, S.; Hessler, N.; Kralisch, D.; Wesarg, F.; Müller, F.A.; Fischer, D.; Hipler, U.-C. Antimicrobial functionalization of bacterial nanocellulose by loading with polihexanide and povidone-iodine. J. Mater. Sci. Mater. Med. 2015, 26, 245. [Google Scholar] [CrossRef]
  189. Okabe, K.; Kimura, H.; Okabe, J.; Kato, A.; Shimizu, H.; Ueda, T.; Shimada, S.; Ogura, Y. Effect of Benzalkonium Chloride on Transscleral Drug Delivery. Investig. Ophthalmol. Vis. Sci. 2005, 46, 703–708. [Google Scholar] [CrossRef] [Green Version]
  190. Johannsdottir, S.; Jansook, P.; Stefansson, E.; Kristinsdottir, I.M.; Asgrimsdottir, G.M.; Loftsson, T. Topical drug delivery to the posterior segment of the eye: The effect of benzalkonium chloride on topical dexamethasone penetration into the eye in vivo. J. Drug Deliv. Sci. Technol. 2018, 48, 125–127. [Google Scholar] [CrossRef]
  191. Lam, H.T.; Le-Vinh, B.; Phan, T.N.Q.; Bernkop-Schnürch, A. Self-emulsifying drug delivery systems and cationic surfactants: Do they potentiate each other in cytotoxicity? J. Pharm. Pharmacol. 2019, 71, 156–166. [Google Scholar] [CrossRef]
  192. Garcia-Fernandez, M.J.; Brackman, G.; Coenye, T.; Concheiro, A.; Alvarez-Lorenzo, C. Antiseptic cyclodextrin-functionalized hydrogels and gauzes for loading and delivery of benzalkonium chloride. Biofouling 2013, 29, 261–271. [Google Scholar] [CrossRef] [PubMed]
  193. Rabea, E.I.; Badawy, M.E.-T.; Stevens, C.V.; Smagghe, G.; Steurbaut, W. Chitosan as antimicrobial agent: Applications and mode of action. Biomacromolecules 2003, 4, 1457–1465. [Google Scholar] [CrossRef] [PubMed]
  194. Abral, H.; Ariksa, J.; Mahardika, M.; Handayani, D.; Aminah, I.; Sandrawati, N.; Pratama, A.B.; Fajri, N.; Sapuan, S.M.; Ilyas, R.A. Transparent and antimicrobial cellulose film from ginger nanofiber. Food Hydrocoll. 2020, 98, 105266. [Google Scholar] [CrossRef]
  195. Zhang, M.; Xiao, B.; Wang, H.; Han, M.K.; Zhang, Z.; Viennois, E.; Xu, C.; Merlin, D. Edible Ginger-derived Nano-lipids Loaded with Doxorubicin as a Novel Drug-delivery Approach for Colon Cancer Therapy. Mol. Ther. 2016, 24, 1783–1796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Baskar, V.; Selvakumar, K. Study on Improving Bioavailability ratio of anti-inflammatory compound from ginger through nano transdermal drug delivery. Asian J. Pharm. Amp Clin. Res. 2012, 5, 241–246. [Google Scholar]
  197. Naghsh, F. Nano drug delivery Study of Anticancer Properties on Ginger using QM/MM Methods. Orient. J. Chem. 2015, 31, 465–478. [Google Scholar] [CrossRef] [Green Version]
  198. Helander, I.M.; Nurmiaho-Lassila, E.-L.; Ahvenainen, R.; Rhoades, J.; Roller, S. Chitosan disrupts the barrier properties of the outer membrane of Gram-negative bacteria. Int. J. Food Microbiol. 2001, 71, 235–244. [Google Scholar] [CrossRef]
  199. Younes, I.; Rinaudo, M. Chitin and Chitosan Preparation from Marine Sources. Structure, Properties and Applications. Mar. Drugs 2015, 13, 1133–1174. [Google Scholar] [CrossRef] [Green Version]
  200. Agnihotri, S.A.; Mallikarjuna, N.N.; Aminabhavi, T.M. Recent advances on chitosan-based micro- and nanoparticles in drug delivery. J. Control. Release 2004, 100, 5–28. [Google Scholar] [CrossRef]
  201. Sannan, T.; Kurita, K.; Iwakura, Y. Studies on chitin, 2. Effect of deacetylation on solubility. Die Makromol. Chem. 1976, 177, 3589–3600. [Google Scholar] [CrossRef]
  202. Rinaudo, M. Chitin and chitosan: Properties and applications. Prog. Polym. Sci. 2006, 31, 603–632. [Google Scholar] [CrossRef]
  203. Ahsan, S.M.; Thomas, M.; Reddy, K.K.; Sooraparaju, S.G.; Asthana, A.; Bhatnagar, I. Chitosan as biomaterial in drug delivery and tissue engineering. Int. J. Biol. Macromol. 2018, 110, 97–109. [Google Scholar] [CrossRef]
  204. Ahmed, T.A.; Aljaeid, B.M. Preparation, characterization, and potential application of chitosan, chitosan derivatives, and chitosan metal nanoparticles in pharmaceutical drug delivery. Drug Des. Dev. Ther. 2016, 10, 483–507. [Google Scholar] [CrossRef] [Green Version]
  205. Mohammed, M.A.; Syeda, J.T.M.; Wasan, K.M.; Wasan, E.K. An Overview of Chitosan Nanoparticles and Its Application in Non-Parenteral Drug Delivery. Pharmaceutics 2017, 9, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Hamedi, H.; Moradi, S.; Hudson, S.M.; Tonelli, A.E. Chitosan based hydrogels and their applications for drug delivery in wound dressings: A review. Carbohydr. Polym. 2018, 199, 445–460. [Google Scholar] [CrossRef] [PubMed]
  207. Ways, T.M.M.; Lau, W.M.; Khutoryanskiy, V.V. Chitosan and Its Derivatives for Application in Mucoadhesive Drug Delivery Systems. Polymers 2018, 10, 267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Quiñones, J.P.; Peniche, H.; Peniche, C. Chitosan Based Self-Assembled Nanoparticles in Drug Delivery. Polymers 2018, 10, 235. [Google Scholar] [CrossRef] [Green Version]
  209. Ludwig, A. The use of mucoadhesive polymers in ocular drug delivery. Adv. Drug Deliv. Rev. 2005, 57, 1595–1639. [Google Scholar] [CrossRef]
  210. Wang, W.; Meng, Q.; Li, Q.; Liu, J.; Zhou, M.; Jin, Z.; Zhao, K. Chitosan Derivatives and Their Application in Biomedicine. Int. J. Mol. Sci. 2020, 21, 487. [Google Scholar] [CrossRef] [Green Version]
  211. Jayakumar, R.; Prabaharan, M.; Sudheesh Kumar, P.T.; Nair, S.V.; Tamura, H. Biomaterials based on chitin and chitosan in wound dressing applications. Biotechnol. Adv. 2011, 29, 322–337. [Google Scholar] [CrossRef]
  212. Kravanja, G.; Primožič, M.; Knez, Ž.; Leitgeb, M. Chitosan-based (Nano)materials for Novel Biomedical Applications. Molecules 2019, 24, 1960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Kaliva, M.; Georgopoulou, A.; Dragatogiannis, D.A.; Charitidis, C.A.; Chatzinikolaidou, M.; Vamvakaki, M. Biodegradable Chitosan-graft-Poly(l-lactide) Copolymers For Bone Tissue Engineering. Polymers 2020, 12, 316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Felt, O.; Buri, P.; Gurny, R. Chitosan: A unique polysaccharide for drug delivery. Drug Dev. Ind. Pharm. 1998, 24, 979–993. [Google Scholar] [CrossRef] [PubMed]
  215. Parhi, R. Drug delivery applications of chitin and chitosan: A review. Environ. Chem. Lett. 2020, 18, 577–594. [Google Scholar] [CrossRef]
  216. Naskar, S.; Kuotsu, K.; Sharma, S. Chitosan-based nanoparticles as drug delivery systems: A review on two decades of research. J. Drug Target. 2019, 27, 379–393. [Google Scholar] [CrossRef]
  217. Tome, L.C.; Fernandes, S.C.M.; Perez, D.S.; Sadocco, P.; Silvestre, A.J.D.; Pascoal Neto, C.; Marrucho, I.M.; Freire, C.S.R. The role of nanocellulose fibers, starch and chitosan on multipolysaccharide based films. Cellulose 2013, 20, 1807–1818. [Google Scholar] [CrossRef]
  218. Devlieghere, F.; Vermeulen, A.; Debevere, J. Chitosan: Antimicrobial activity, interactions with food components and applicability as a coating on fruit and vegetables. Food Microbiol. 2004, 21, 703–714. [Google Scholar] [CrossRef]
  219. Goy, R.C.; de Britto, D.; Assis, O.B.G. A review of the antimicrobial activity of chitosan. Polímeros 2009, 19, 241–247. [Google Scholar] [CrossRef]
  220. Zhang, P.; Chen, L.; Zhang, Q.; Hong, F.F. Using In situ Dynamic Cultures to Rapidly Biofabricate Fabric-Reinforced Composites of Chitosan/Bacterial Nanocellulose for Antibacterial Wound Dressings. Front. Microbiol. 2016, 7, 260. [Google Scholar] [CrossRef] [Green Version]
  221. Sampath, U.G.T.M.; Ching, Y.C.; Chuah, C.H.; Singh, R.; Lin, P.-C. Preparation and characterization of nanocellulose reinforced semi-interpenetrating polymer network of chitosan hydrogel. Cellulose 2017, 24, 2215–2228. [Google Scholar] [CrossRef]
  222. Poonguzhali, R.; Basha, S.K.; Kumari, V.S. Synthesis and characterization of chitosan-PVP-nanocellulose composites for in-vitro wound dressing application. Int. J. Biol. Macromol. 2017, 105, 111–120. [Google Scholar] [CrossRef] [PubMed]
  223. Kumar, G.P.; Phani, A.R.; Prasad, R.G.S.V.; Sanganal, J.S.; Manali, N.; Gupta, R.; Rashmi, N.; Prabhakara, G.S.; Salins, C.P.; Sandeep, K.; et al. Polyvinylpyrrolidone oral films of enrofloxacin: Film characterization and drug release. Int. J. Pharm. 2014, 471, 146–152. [Google Scholar] [CrossRef] [PubMed]
  224. Kadajji, V.G.; Betageri, G.V. Water Soluble Polymers for Pharmaceutical Applications. Polymers 2011, 3, 1972–2009. [Google Scholar] [CrossRef] [Green Version]
  225. Wu, H.-D.; Wu, I.-D.; Chang, F.-C. The interaction behavior of polymer electrolytes composed of poly(vinyl pyrrolidone) and lithium perchlorate (LiClO4). Polymer 2001, 42, 555–562. [Google Scholar] [CrossRef]
  226. Hasan, A.; Waibhaw, G.; Tiwari, S.; Dharmalingam, K.; Shukla, I.; Pandey, L.M. Fabrication and characterization of chitosan, polyvinylpyrrolidone, and cellulose nanowhiskers nanocomposite films for wound healing drug delivery application. J. Biomed. Mater. Res. Part A 2017, 105, 2391–2404. [Google Scholar] [CrossRef]
  227. Moore, A. Antimicrobial Tissue Paper Could Help Fight the Spread of COVID-19. College of Natural Resources News, 30 March 2020. [Google Scholar]
  228. Patel, M. Nanoparticle-based Antimicrobial Paper as Spread-breaker for Coronavirus. Pap. Technol. Int. 2020, 62, 20–25. [Google Scholar]
  229. Tyagi, P.; Mathew, R.; Opperman, C.; Jameel, H.; Gonzalez, R.; Lucia, L.; Hubbe, M.; Pal, L. High-Strength Antibacterial Chitosan-Cellulose Nanocrystal Composite Tissue Paper. Langmuir 2019, 35, 104–112. [Google Scholar] [CrossRef]
  230. Patel, M. Antimicrobial Paper Embedded with Nanoparticles as Spread-Breaker for Corona Virus. J. Environ. Life Sci. 2020, 6, 001–012. [Google Scholar]
  231. Sundaram, J.; Pant, J.; Goudie, M.J.; Mani, S.; Handa, H. Antimicrobial and Physicochemical Characterization of Biodegradable, Nitric Oxide-Releasing Nanocellulose-Chitosan Packaging Membranes. J. Agric. Food Chem. 2016, 64, 5260–5266. [Google Scholar] [CrossRef]
  232. Fang, F.C. Antimicrobial actions of nitric oxide. Nitric Oxide 2012, 27, S10. [Google Scholar] [CrossRef]
  233. Brisbois, E.J.; Kim, M.; Wang, X.; Mohammed, A.; Major, T.C.; Wu, J.; Brownstein, J.; Xi, C.; Handa, H.; Bartlett, R.H.; et al. Improved Hemocompatibility of Multilumen Catheters via Nitric Oxide (NO) Release from S-Nitroso-N-acetylpenicillamine (SNAP) Composite Filled Lumen. ACS Appl. Mater. Interfaces 2016, 8, 29270–29279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Sogut, E.; Seydim, A.C. Development of Chitosan and Polycaprolactone based active bilayer films enhanced with nanocellulose and grape seed extract. Carbohydr. Polym. 2018, 195, 180–188. [Google Scholar] [CrossRef] [PubMed]
  235. Furiga, A.; Lonvaud-Funel, A.; Badet, C. In vitro study of antioxidant capacity and antibacterial activity on oral anaerobes of a grape seed extract. Food Chem. 2009, 113, 1037–1040. [Google Scholar] [CrossRef]
  236. Benoit, M.-A.; Baras, B.; Gillard, J. Preparation and characterization of protein-loaded poly(ε-caprolactone) microparticles for oral vaccine delivery. Int. J. Pharm. 1999, 184, 73–84. [Google Scholar] [CrossRef]
  237. Pitt, C.G.; Jeffcoat, A.R.; Zweidinger, R.A.; Schindler, A. Sustained drug delivery systems. I. The permeability of poly(ϵ-caprolactone), poly(DL-lactic acid), and their copolymers. J. Biomed. Mater. Res. 1979, 13, 497–507. [Google Scholar] [CrossRef]
  238. Sahoo, S.; Sasmal, A.; Nanda, R.; Phani, A.R.; Nayak, P.L. Synthesis of chitosan–polycaprolactone blend for control delivery of ofloxacin drug. Carbohydr. Polym. 2010, 79, 106–113. [Google Scholar] [CrossRef]
  239. Roemhild, K.; Wiegand, C.; Hipler, U.-C.; Heinze, T. Novel bioactive amino-functionalized cellulose nanofibers. Macromol. Rapid Commun. 2013, 34, 1767–1771. [Google Scholar] [CrossRef]
  240. Saini, S.; Belgacem, M.N.; Salon, M.-C.B.; Bras, J. Non leaching biomimetic antimicrobial surfaces via surface functionalisation of cellulose nanofibers with aminosilane. Cellulose 2016, 23, 795–810. [Google Scholar] [CrossRef]
  241. He, W.; Zhang, Z.; Zheng, Y.; Qiao, S.; Xie, Y.; Sun, Y.; Qiao, K.; Feng, Z.; Wang, X.; Wang, J. Preparation of aminoalkyl-grafted bacterial cellulose membranes with improved antimicrobial properties for biomedical applications. J. Biomed. Mater. Res. A 2020, 108, 1086–1098. [Google Scholar] [CrossRef]
  242. Weishaupt, R.; Heuberger, L.; Siqueira, G.; Gutt, B.; Zimmermann, T.; Maniura-Weber, K.; Salentinig, S.; Faccio, G. Enhanced Antimicrobial Activity and Structural Transitions of a Nanofibrillated Cellulose–Nisin Biocomposite Suspension. ACS Appl. Mater. Interfaces 2018, 10, 20170–20181. [Google Scholar] [CrossRef]
  243. dos Santos, C.A.; dos Santos, G.R.; Soeiro, V.S.; dos Santos, J.R.; de Araujo Rebelo, M.; Chaud, M.V.; Gerenutti, M.; Grotto, D.; Pandit, R.; Rai, M.; et al. Bacterial nanocellulose membranes combined with nisin: A strategy to prevent microbial growth. Cellulose 2018, 25, 6681–6689. [Google Scholar] [CrossRef]
  244. Ugurlu, T.; Turkoglu, M.; Gurer, U.S.; Akarsu, B.G. Colonic delivery of compression coated nisin tablets using pectin/HPMC polymer mixture. Eur. J. Pharm. Biopharm. 2007, 67, 202–210. [Google Scholar] [CrossRef] [PubMed]
  245. Correia, R.C.; Jozala, A.F.; Martins, K.F.; Penna, T.C.V.; de Rezende Duek, E.A.; de Oliveira Rangel-Yagui, C.; Lopes, A.M. Poly(lactic-co-glycolic acid) matrix incorporated with nisin as a novel antimicrobial biomaterial. World J. Microbiol. Biotechnol. 2015, 31, 649–659. [Google Scholar] [CrossRef] [PubMed]
  246. Shin, J.M.; Gwak, J.W.; Kamarajan, P.; Fenno, J.C.; Rickard, A.H.; Kapila, Y.L. Biomedical applications of nisin. J. Appl. Microbiol. 2016, 120, 1449–1465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Maurer, H.R. Bromelain: Biochemistry, pharmacology and medical use. Cell. Mol. Life Sci. 2001, 58, 1234–1245. [Google Scholar] [CrossRef] [PubMed]
  248. Cherian, B.M.; Leão, A.L.; de Souza, S.F.; Thomas, S.; Pothan, L.A.; Kottaisamy, M. Isolation of nanocellulose from pineapple leaf fibres by steam explosion. Carbohydr. Polym. 2010, 81, 720–725. [Google Scholar] [CrossRef]
  249. Ataide, J.A.; de Carvalho, N.M.; de Araújo Rebelo, M.; Chaud, M.V.; Grotto, D.; Gerenutti, M.; Rai, M.; Mazzola, P.G.; Jozala, A.F. Bacterial Nanocellulose Loaded with Bromelain: Assessment of Antimicrobial, Antioxidant and Physical-Chemical Properties. Sci Rep. 2017, 7, 18031. [Google Scholar] [CrossRef]
  250. Bagga, P.; Ansari, T.M.; Siddiqui, H.H.; Syed, A.; Bahkali, A.H.; Rahman, M.A.; Khan, M.S. Bromelain capped gold nanoparticles as the novel drug delivery carriers to aggrandize effect of the antibiotic levofloxacin. EXCLI J. 2016, 15, 772–780. [Google Scholar] [CrossRef]
  251. Nasiri, R.; Hamzehalipour Almaki, J.; Idris, A.; Nasiri, M.; Irfan, M.; Majid, F.A.A.; Rashidi Nodeh, H.; Hasham, R. Targeted delivery of bromelain using dual mode nanoparticles: Synthesis, physicochemical characterization, in vitro and in vivo evaluation. RSC Adv. 2017, 7, 40074–40094. [Google Scholar] [CrossRef] [Green Version]
  252. Bhatnagar, P.; Patnaik, S.; Srivastava, A.K.; Mudiam, M.K.R.; Shukla, Y.; Panda, A.K.; Pant, A.B.; Kumar, P.; Gupta, K.C. Anti-cancer activity of bromelain nanoparticles by oral administration. J. Biomed. Nanotechnol. 2014, 10, 3558–3575. [Google Scholar] [CrossRef]
  253. Ahluwalia, A.K.; Sekhon, B.S. Enzybiotics: A promising approach to fight infectious diseases and an upcoming need for future. J. Pharm. Educ. Res. 2012, 3, 42–51. [Google Scholar]
  254. Sampaio, L.M.P.; Padrao, J.; Faria, J.; Silva, J.P.; Silva, C.J.; Dourado, F.; Zille, A. Laccase immobilization on bacterial nanocellulose membranes: Antimicrobial, kinetic and stability properties. Carbohydr. Polym. 2016, 145, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Zhang, X.; Chen, T.; Lim, J.; Gu, F.; Fang, F.; Cheng, L.; Campanella, O.H.; Hamaker, B.R. Acid gelation of soluble laccase-crosslinked corn bran arabinoxylan and possible gel formation mechanism. Food Hydrocoll. 2019, 92, 1–9. [Google Scholar] [CrossRef]
  256. Raghavendra, G.M.; Jayaramudu, T.; Varaprasad, K.; Ramesh, S.; Raju, K.M. Microbial resistant nanocurcumin-gelatin-cellulose fibers for advanced medical applications. RSC Adv. 2013, 4, 3494–3501. [Google Scholar] [CrossRef]
  257. Aggarwal, B.B.; Sundaram, C.; Malani, N.; Ichikawa, H. Curcumin: The Indian solid gold. In The Molecular Targets and Therapeutic Uses of Curcumin in Health and Disease; Aggarwal, B.B., Surh, Y.-J., Shishodia, S., Eds.; Advances in Experimental Medicine and Biology; Springer: Boston, MA, USA, 2007; pp. 1–75. ISBN 978-0-387-46401-5. [Google Scholar]
  258. Shaikh, J.; Ankola, D.D.; Beniwal, V.; Singh, D.; Kumar, M.N.V.R. Nanoparticle encapsulation improves oral bioavailability of curcumin by at least 9-fold when compared to curcumin administered with piperine as absorption enhancer. Eur. J. Pharm. Sci. 2009, 37, 223–230. [Google Scholar] [CrossRef]
  259. Sanoj Rejinold, N.; Sreerekha, P.R.; Chennazhi, K.P.; Nair, S.V.; Jayakumar, R. Biocompatible, biodegradable and thermo-sensitive chitosan-g-poly (N-isopropylacrylamide) nanocarrier for curcumin drug delivery. Int. J. Biol. Macromol. 2011, 49, 161–172. [Google Scholar] [CrossRef]
  260. Sun, D.; Zhuang, X.; Xiang, X.; Liu, Y.; Zhang, S.; Liu, C.; Barnes, S.; Grizzle, W.; Miller, D.; Zhang, H.G. A Novel Nanoparticle Drug Delivery System: The Anti-inflammatory Activity of Curcumin Is Enhanced When Encapsulated in Exosomes. Mol. Ther. 2010, 18, 1606–1614. [Google Scholar] [CrossRef]
  261. Pham, X.N.; Nguyen, T.P.; Pham, T.N.; Tran, T.T.N.; Tran, T.V.T. Synthesis and characterization of chitosan-coated magnetite nanoparticles and their application in curcumin drug delivery. Adv. Nat. Sci Nanosci. Nanotechnol. 2016, 7, 045010. [Google Scholar] [CrossRef]
  262. Gupta, M.; Chauhan, D.N.; Sharma, V.; Chauhan, N.S. Novel Drug Delivery Systems for Phytoconstituents; CRC Press: Boca Raton, FL, USA, 2019; ISBN 978-1-351-05762-2. [Google Scholar]
  263. Sipponen, A.; Laitinen, K. Antimicrobial properties of natural coniferous rosin in the European Pharmacopoeia challenge test. APMIS 2011, 119, 720–724. [Google Scholar] [CrossRef]
  264. De Castro, D.O.; Bras, J.; Gandini, A.; Belgacem, N. Surface grafting of cellulose nanocrystals with natural antimicrobial rosin mixture using a green process. Carbohydr. Polym. 2016, 137, 1–8. [Google Scholar] [CrossRef]
  265. El-Sayed Abdel-Raouf, M.; Mahmoud Abdul-Raheim, A.-R. Rosin: Chemistry, Derivatives, and Applications: A review. BAOJ Chem. 2018, 4, 039. [Google Scholar]
  266. Yadav, B.K.; Gidwani, B.; Vyas, A. Rosin: Recent advances and potential applications in novel drug delivery system. J. Bioact. Compat. Polym. 2016, 31, 111–126. [Google Scholar] [CrossRef]
  267. Uddin, K.M.A.; Orelma, H.; Mohammadi, P.; Borghei, M.; Laine, J.; Linder, M.; Rojas, O.J. Retention of lysozyme activity by physical immobilization in nanocellulose aerogels and antibacterial effects. Cellulose 2017, 24, 2837–2848. [Google Scholar] [CrossRef]
  268. Barbiroli, A.; Bonomi, F.; Capretti, G.; Iametti, S.; Manzoni, M.; Piergiovanni, L.; Rollini, M. Antimicrobial activity of lysozyme and lactoferrin incorporated in cellulose-based food packaging. Food Control 2012, 26, 387–392. [Google Scholar] [CrossRef] [Green Version]
  269. Abeyrathne, E.D.N.S.; Lee, H.Y.; Ahn, D.U. Egg white proteins and their potential use in food processing or as nutraceutical and pharmaceutical agents—A review. Poult. Sci. 2013, 92, 3292–3299. [Google Scholar] [CrossRef]
  270. Alves, M.; Vieira, N.S.M.; Rebelo, L.P.N.; Araújo, J.M.M.; Pereiro, A.B.; Archer, M. Fluorinated ionic liquids for protein drug delivery systems: Investigating their impact on the structure and function of lysozyme. Int. J. Pharm. 2017, 526, 309–320. [Google Scholar] [CrossRef]
  271. Wu, T.; Huang, J.; Jiang, Y.; Hu, Y.; Ye, X.; Liu, D.; Chen, J. Formation of hydrogels based on chitosan/alginate for the delivery of lysozyme and their antibacterial activity. Food Chem. 2018, 240, 361–369. [Google Scholar] [CrossRef]
  272. Marchese, A.; Barbieri, R.; Sanches-Silva, A.; Daglia, M.; Nabavi, S.F.; Jafari, N.J.; Izadi, M.; Ajami, M.; Nabavi, S.M. Antifungal and antibacterial activities of allicin: A review. Trends Food Sci. Technol. 2016, 52, 49–56. [Google Scholar] [CrossRef]
  273. Jamindar, D.; Patidar, N.; Jain, S. Formulation and characterization of allicin-amphotericin-b liposomal gel for the treatment of fungal infections. J. Drug Deliv. Ther. 2017, 7, 69–70. [Google Scholar] [CrossRef]
  274. Jebali, A.; Hekmatimoghaddam, S.; Behzadi, A.; Rezapor, I.; Mohammadi, B.H.; Jasemizad, T.; Yasini, S.A.; Javadzadeh, M.; Amiri, A.; Soltani, M.; et al. Antimicrobial activity of nanocellulose conjugated with allicin and lysozyme. Cellulose 2013, 20, 2897–2907. [Google Scholar] [CrossRef]
  275. Jafary, R.; Mehrizi, M.K.; Hekmatimoghaddam, S.H.; Jebali, A. Antibacterial property of cellulose fabric finished by allicin-conjugated nanocellulose. J. Text. Inst. 2015, 106, 683–689. [Google Scholar] [CrossRef]
  276. Atef, M.; Rezaei, M.; Behrooz, R. Characterization of physical, mechanical, and antibacterial properties of agar-cellulose bionanocomposite films incorporated with savory essential oil. Food Hydrocoll. 2015, 45, 150–157. [Google Scholar] [CrossRef]
  277. Deans, S.G.; Svoboda, K.P. Antibacterial activity of summer savory (Satureja hortensis L) essential oil and its constituents. J. Hortic. Sci. 1989, 64, 205–210. [Google Scholar] [CrossRef]
  278. Feyzioglu, G.C.; Tornuk, F. Development of chitosan nanoparticles loaded with summer savory (Satureja hortensis L.) essential oil for antimicrobial and antioxidant delivery applications. LWT 2016, 70, 104–110. [Google Scholar] [CrossRef]
  279. Wojtyczka, R.D.; Kępa, M.; Idzik, D.; Kubina, R.; Kabała-Dzik, A.; Dziedzic, A.; Wąsik, T.J. In Vitro Antimicrobial Activity of Ethanolic Extract of Polish Propolis against Biofilm Forming Staphylococcus epidermidis Strains. Evid.Based Complement. Alternat. Med. 2013, 2013. [Google Scholar] [CrossRef] [Green Version]
  280. da Silva Barud, H.; de Araújo Júnior, A.M.; Saska, S.; Mestieri, L.B.; Campos, J.A.D.B.; de Freitas, R.M.; Ferreira, N.U.; Nascimento, A.P.; Miguel, F.G.; de Oliveira Lima Leite Vaz, M.M.; et al. Antimicrobial Brazilian Propolis (EPP-AF) Containing Biocellulose Membranes as Promising Biomaterial for Skin Wound Healing. Evid. Based Complement. Alternat. Med. 2013, 2013. [Google Scholar] [CrossRef]
  281. Al-Waili, N.; Al-Ghamdi, A.; Ansari, M.J.; Al-Attal, Y.; Salom, K. Synergistic effects of honey and propolis toward drug multi-resistant Staphylococcus aureus, Escherichia coli and Candida albicans isolates in single and polymicrobial cultures. Int. J. Med. Sci. 2012, 9, 793–800. [Google Scholar] [CrossRef] [Green Version]
  282. Dantas, T.N.C.; Silva, H.S.R.C.; Dantas Neto, A.A.; Marcucci, M.C.; Maciel, M.A.M. Development of a new propolis microemulsion system for topical applications. Rev. Bras. Farmacogn. 2010, 20, 368–375. [Google Scholar] [CrossRef] [Green Version]
  283. Ahmed, E.T.; Abo-Salem, O.M.; Osman, A. The Influence of Egyptian Propolis on Induced Burn Wound Healing in Diabetic Rats; Antibacterial Mechanism. Sci. J. Med. Clin. Trials 2011, 1. [Google Scholar] [CrossRef]
  284. Berretta, A.A.; Nascimento, A.P.; Bueno, P.C.P.; de Oliveira Lima Leite Vaz, M.M.; Marchetti, J.M. Propolis Standardized Extract (EPP-AF®), an Innovative Chemically and Biologically Reproducible Pharmaceutical Compound for Treating Wounds. Int. J. Biol. Sci. 2012, 8, 512–521. [Google Scholar] [CrossRef]
  285. Balata, G.; Nahas, H.M.E.; Radwan, S. Propolis organogel as a novel topical delivery system for treating wounds. Drug Deliv. 2014, 21, 55–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  286. Rassu, G.; Cossu, M.; Langasco, R.; Carta, A.; Cavalli, R.; Giunchedi, P.; Gavini, E. Propolis as lipid bioactive nano-carrier for topical nasal drug delivery. Colloids Surf. B Biointerfaces 2015, 136, 908–917. [Google Scholar] [CrossRef] [PubMed]
  287. Yang, X.; Fan, L.; Ma, L.; Wang, Y.; Lin, S.; Yu, F.; Pan, X.; Luo, G.; Zhang, D.; Wang, H. Green electrospun Manuka honey/silk fibroin fibrous matrices as potential wound dressing. Mater. Des. 2017, 119, 76–84. [Google Scholar] [CrossRef]
  288. Boateng, J.S.; Matthews, K.H.; Stevens, H.N.E.; Eccleston, G.M. Wound Healing Dressings and Drug Delivery Systems: A Review. J. Pharm. Sci. 2008, 97, 2892–2923. [Google Scholar] [CrossRef] [PubMed]
  289. Matthews, K.H. 14 - Drug delivery dressings. In Advanced Wound Repair Therapies; Farrar, D., Ed.; Woodhead Publishing Series in Biomaterials; Woodhead Publishing: Cambridge, UK, 2011; pp. 361–394. ISBN 978-1-84569-700-6. [Google Scholar]
  290. Tenci, M.; Rossi, S.; Bonferoni, M.C.; Sandri, G.; Boselli, C.; Di Lorenzo, A.; Daglia, M.; Icaro Cornaglia, A.; Gioglio, L.; Perotti, C.; et al. Particulate systems based on pectin/chitosan association for the delivery of manuka honey components and platelet lysate in chronic skin ulcers. Int. J. Pharm. 2016, 509, 59–70. [Google Scholar] [CrossRef] [PubMed]
  291. Subash Babu, P.; Prabuseenivasan, S.; Ignacimuthu, S. Cinnamaldehyde—A potential antidiabetic agent. Phytomedicine 2007, 14, 15–22. [Google Scholar] [CrossRef] [PubMed]
  292. Devi, K.P.; Nisha, S.A.; Sakthivel, R.; Pandian, S.K. Eugenol (an essential oil of clove) acts as an antibacterial agent against Salmonella typhi by disrupting the cellular membrane. J. Ethnopharmacol. 2010, 130, 107–115. [Google Scholar] [CrossRef] [PubMed]
  293. Sanla-Ead, N.; Jangchud, A.; Chonhenchob, V.; Suppakul, P. Antimicrobial Activity of Cinnamaldehyde and Eugenol and Their Activity after Incorporation into Cellulose-based Packaging Films. Packag. Technol. Sci. 2012, 25, 7–17. [Google Scholar] [CrossRef]
  294. Kenawy, E.; Omer, A.M.; Tamer, T.M.; Elmeligy, M.A.; Eldin, M.S.M. Fabrication of biodegradable gelatin/chitosan/cinnamaldehyde crosslinked membranes for antibacterial wound dressing applications. Int. J. Biol. Macromol. 2019, 139, 440–448. [Google Scholar] [CrossRef]
  295. Bang, K.-H.; Kim, K.S. Development of trans-cinnamaldehyde self-microemulsifying drug delivery system (SMEDDS) with superior stability. J. Korea Acad.-Ind. Coop. Soc. 2019, 20, 555–562. [Google Scholar] [CrossRef]
  296. Patole, V.C.; Chaudhari, S.P.; Pandit, A.P.; Lokhande, P.P. Thymol and eugenol loaded chitosan dental film for treatment of periodontitis. Indian Drugs 2019, 54, 51–58. [Google Scholar]
  297. Esmaeili, F.; Rajabnejhad, S.; Partoazar, A.R.; Mehr, S.E.; Faridi-Majidi, R.; Sahebgharani, M.; Syedmoradi, L.; Rajabnejhad, M.R.; Amani, A. Anti-inflammatory effects of eugenol nanoemulsion as a topical delivery system. Pharm. Dev. Technol. 2016, 21, 887–893. [Google Scholar] [CrossRef] [PubMed]
  298. El Khayat, N.W.; Donia, A.A.; Mady, O.Y.; El Maghraby, G.M. Optimization of eugenol microemulsion for transdermal delivery of indomethacin. J. Drug Deliv. Sci. Technol. 2018, 48, 311–318. [Google Scholar] [CrossRef]
  299. Silva, S.S.; Caridade, S.G.; Mano, J.F.; Reis, R.L. Effect of crosslinking in chitosan/aloe vera-based membranes for biomedical applications. Carbohydr. Polym. 2013, 98, 581–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  300. Godinho, J.F.; Berti, F.V.; Müller, D.; Rambo, C.R.; Porto, L.M. Incorporation of Aloe vera extracts into nanocellulose during biosynthesis. Cellulose 2016, 23, 545–555. [Google Scholar] [CrossRef]
  301. Saibuatong, O.; Phisalaphong, M. Novo aloe vera–bacterial cellulose composite film from biosynthesis. Carbohydr. Polym. 2010, 79, 455–460. [Google Scholar] [CrossRef]
  302. Gupta, B.; Agarwal, R.; Alam, S. Aloe Vera Loaded Poly(Vinyl Alcohol)–Poly(Ethylene Oxide)-Carboxymethyl Cellulose-Polyester Nonwoven Membranes. J. Biomater. Tissue Eng. 2013, 3, 503–511. [Google Scholar] [CrossRef]
  303. Laux, A.; Gouws, C.; Hamman, J.H. Aloe vera gel and whole leaf extract: Functional and versatile excipients for drug delivery? Expert Opin. Drug Deliv. 2019, 16, 1283–1285. [Google Scholar] [CrossRef] [Green Version]
  304. Jacob, J.; Haponiuk, J.T.; Thomas, S.; Peter, G.; Gopi, S. Use of Ginger Nanofibers for the Preparation of Cellulose Nanocomposites and Their Antimicrobial Activities. Fibers 2018, 6, 79. [Google Scholar] [CrossRef] [Green Version]
  305. Jacob, J.; Peter, G.; Thomas, S.; Haponiuk, J.T.; Gopi, S. Chitosan and polyvinyl alcohol nanocomposites with cellulose nanofibers from ginger rhizomes and its antimicrobial activities. Int. J. Biol. Macromol. 2019, 129, 370–376. [Google Scholar] [CrossRef]
  306. Dehnad, D.; Mirzaei, H.; Emam-Djomeh, Z.; Jafari, S.-M.; Dadashi, S. Thermal and antimicrobial properties of chitosan–nanocellulose films for extending shelf life of ground meat. Carbohydr. Polym. 2014, 109, 148–154. [Google Scholar] [CrossRef] [PubMed]
  307. Mujtaba, M.; Salaberria, A.M.; Andres, M.A.; Kaya, M.; Gunyakti, A.; Labidi, J. Utilization of flax (Linum usitatissimum) cellulose nanocrystals as reinforcing material for chitosan films. Int. J. Biol. Macromol. 2017, 104, 944–952. [Google Scholar] [CrossRef] [PubMed]
  308. Saini, S.; Sillard, C.; Belgacem, M.N.; Bras, J. Nisin anchored cellulose nanofibers for long term antimicrobial active food packaging. RSC Adv. 2016, 6, 12422–12430. [Google Scholar] [CrossRef]
  309. Salmieri, S.; Islam, F.; Khan, R.A.; Hossain, F.M.; Ibrahim, H.M.M.; Miao, C.; Hamad, W.Y.; Lacroix, M. Antimicrobial nanocomposite films made of poly(lactic acid)-cellulose nanocrystals (PLA-CNC) in food applications: Part A—Effect of nisin release on the inactivation of Listeria monocytogenes in ham. Cellulose 2014, 21, 1837–1850. [Google Scholar] [CrossRef]
  310. Tangsatianpan, V.; Torgbo, S.; Sukyai, P. Release Kinetic Model and Antimicrobial Activity of Freeze-Dried Curcumin-loaded Bacterial Nanocellulose Composite. Polym. Sci. Ser. A 2020. [Google Scholar] [CrossRef]
  311. de Castro, D.O.; Tabary, N.; Martel, B.; Gandini, A.; Belgacem, N.; Bras, J. Controlled release of carvacrol and curcumin: Bio-based food packaging by synergism action of TEMPO-oxidized cellulose nanocrystals and cyclodextrin. Cellulose 2018, 25, 1249–1263. [Google Scholar] [CrossRef]
  312. Niu, X.; Liu, Y.; Song, Y.; Han, J.; Pan, H. Rosin modified cellulose nanofiber as a reinforcing and co-antimicrobial agents in polylactic acid /chitosan composite film for food packaging. Carbohydr. Polym. 2018, 183, 102–109. [Google Scholar] [CrossRef]
  313. Bayazidi, P.; Almasi, H.; Asl, A.K. Immobilization of lysozyme on bacterial cellulose nanofibers: Characteristics, antimicrobial activity and morphological properties. Int. J. Biol. Macromol. 2018, 107, 2544–2551. [Google Scholar] [CrossRef]
Figure 1. Structure of cellobiose that shows the repeated AGUs (summarized by [4]).
Figure 1. Structure of cellobiose that shows the repeated AGUs (summarized by [4]).
Polymers 12 02825 g001
Figure 2. Different sources and types of nanocellulose with representative electron microscope images [8,9,10], reproduced by permission of The Royal Society of Chemistry [11,12,13,14].
Figure 2. Different sources and types of nanocellulose with representative electron microscope images [8,9,10], reproduced by permission of The Royal Society of Chemistry [11,12,13,14].
Polymers 12 02825 g002
Figure 3. Unique properties allow the use of nanocellulose in many Biomedical applications [2].
Figure 3. Unique properties allow the use of nanocellulose in many Biomedical applications [2].
Polymers 12 02825 g003
Figure 4. Preparation procedure of single-membrane and double-membrane microsphere hydrogels [99].
Figure 4. Preparation procedure of single-membrane and double-membrane microsphere hydrogels [99].
Polymers 12 02825 g004
Figure 5. Schematic procedure to prepare hybrids consisting of BNC and AgNPs (summarized by [23]).
Figure 5. Schematic procedure to prepare hybrids consisting of BNC and AgNPs (summarized by [23]).
Polymers 12 02825 g005
Figure 6. Schematic procedure of preparation of chitosan from chitin (summarized by [193,199]).
Figure 6. Schematic procedure of preparation of chitosan from chitin (summarized by [193,199]).
Polymers 12 02825 g006
Figure 7. Schematic procedure for the formation of nanocellulose-chitosan hydrogel (summarized by [221]).
Figure 7. Schematic procedure for the formation of nanocellulose-chitosan hydrogel (summarized by [221]).
Polymers 12 02825 g007
Figure 8. Preparation of chitosan/CNC composite tissue paper that can help prevent the spread of infectious diseases.
Figure 8. Preparation of chitosan/CNC composite tissue paper that can help prevent the spread of infectious diseases.
Polymers 12 02825 g008
Figure 9. General procedure for bacterial cellulose silane chemical grafting with APS (summarized by [73]).
Figure 9. General procedure for bacterial cellulose silane chemical grafting with APS (summarized by [73]).
Polymers 12 02825 g009
Figure 10. Schematic procedure of conjugation between nanocellulose, allicin, and lysozyme (summarized by [274]).
Figure 10. Schematic procedure of conjugation between nanocellulose, allicin, and lysozyme (summarized by [274]).
Polymers 12 02825 g010
Table 1. Diverse CNC-based delivery systems with specific modification agents, carrier forms, model drugs, and potential applications.
Table 1. Diverse CNC-based delivery systems with specific modification agents, carrier forms, model drugs, and potential applications.
Co-CarrierModification AgentCarrier FormModel DrugPossible ApplicationRef.
Sodium alginateCaCl2, EPTMAC, PEIHydrogelCeftazidime hydrate and epidermal growth factor humanCodelivery of drugs in the oral administration and wound dressing[99]
Cyclodextrin-ComplexesCurcuminAnticancer drug delivery systems[100]
Quaternized celluloseCHPTAC, β-GPHydrogelDoxorubicinLocalized and sustained drug delivery depot systems for anticancer therapy[101]
-APTES, PPI-dendrimer,
FA
Lyophilized NPsDoxorubicinDelivery of anticancer drug[102]
-Aln, ApADispersion--Bone therapied and theranostics[103]
Sodium alginate-HydrogelIbuprofenDrug carrier[104]
PVA-Hydrogel lensesChitosan–poly(acrylic acid) NPsOphthalmic use as a drug carrier and as cornea regeneration implant[105]
-PVPFilmHoneyWound dressing for the treatment of chronic wounds[106]
-CTABSuspensionCurcuminDrug carrier for hydrophobic drugs[107]
-PVAFilmCurcuminAntimicrobial drug delivery in a diabetic wound dressing[108]
ChitosanTween 20, GAHydrogelCurcuminDrug delivery of curcumin[110]
-Tween 20SolutionCurcuminDelivery of curcumin to the stomach and upper intestinal tract[111]
FO and PEI-Lyophilized hybridsDoxorubicinLayer-by-layer assembly with lysosomal pH-controlled drug release into the nucleus[112]
PCL-NanofibersTetracyclineDrug delivery system[113]
CaCl2—calcium chloride, EPTMAC—2,3-epoxypropyl trimethylammonium chloride, PEI—polyethylenimine, CHPTAC—3-chloro-2-hydroxypropyltrimethylammonium chloride, β-GP—β-glycerophosphate, PPI-dendrimer—poly(propylene imine) dendrimer, APTES—(3-aminopropyl)triethoxysilane, FA—folic acid, Aln—sodium alendronate, ApA—3-aminoropylphosphoric acid, PVA—polyvinyl alcohol, PVP—polyvinylpyrrolidone, CTAB—cetyl trimethylammonium bromide, GA—glutaraldehyde, FO—folate, PCL—poly(ε-caprolactone).
Table 2. Diverse CNF-based delivery systems with specific modification agents, carrier forms, model drugs, and potential applications.
Table 2. Diverse CNF-based delivery systems with specific modification agents, carrier forms, model drugs, and potential applications.
Co-CarrierModification AgentCarrier FormModel DrugPossible ApplicationRef.
-LADry foams and filmsRiboflavinGastro retentive drug delivery[115]
--Nanopapers and nanofoamsIndomethacinFast and intermediate release profiles for drug delivery[116]
--AerogelsBendamustine hydrochlorideCarriers for oral drug delivery[117]
HFBII-EmulsionNaproxen and ibuprofenDrug delivery applications via the oral route[118]
-GTMACNanofoamFurosemideProlonged drug delivery in the upper part of the gastrointestinal tract[119]
Fe3O4 -Ag2O quantum dots-PowderEtoposide and MethotrexateCarrier of anticancer drugs for skin cancer[120]
-FITIC-DEX, lysozyme, and BSAHydrogelMetronidazole, nadolol, and ketoprofenControlled delivery of several types of molecules[121]
Sodium alginate-Beads (dried hydrogel)Metformin hydrochlorideDrug carrier[122]
Sodium alginate-FilmAmpicillinSupporting material, drug delivery system for Tissue Engineering[123]
PDAcalcium ionHydrogelTetracycline
hydrochloride
Drug delivery vehicle[124]
Chitosan-FilmKetorolac tromethamineTransdermal delivery systems[125]
HPMC-FilmKetorolac tromethamineFood packaging and transdermal drug delivery applications[126]
PLA-HydrogelMetoprolol and nadololModulating sustained drug release[127]
GelatinDialdehyde starchCryogel5-fluorouracilCarrier for controlled drug release[129]
Starch/pectinGlycerinFilmMethotrexateColonic drug release[130]
--AerogelIbuprofen and amoxicillinDrug delivery vehicle[131]
--HydrogelIbuprofenControlled drug release system[132]
LA—lauric acid sodium salt, HFBII—class II hydrophobin protein, GTMAC—glycidyltrimethylammonium chloride, FITIC-DEX—fluoresceinisothiocyanate-dextran, PDA—polydopamine, HPMC—hydroxypropylmethylcellulose, PLA—poly(lactic acid).
Table 3. Diverse BNC-based delivery systems with specific modification agents, carrier forms, model drugs, and potential applications.
Table 3. Diverse BNC-based delivery systems with specific modification agents, carrier forms, model drugs, and potential applications.
Co-CarrierModification AgentCarrier FormModel DrugPossible ApplicationRef.
Starch/pectinGlycerinFilmMethotrexateColonic drug release[130]
Poloxamer-Gel and micelleOctenidineLong-term dermal wound treatment and drug delivery[133]
Collagen-FilmLidocaineBiodegradable microneedles for transdermal drug delivery[134]
PMGlyMGly, AAPH, and MBAMembranesDiclofenacDrug carriers for dermal and oral drug delivery[135]
--HydrogelBSACarrier for controlled delivery of proteins[136]
--ScaffoldsCaco-2-cellsCaco-2-based in vitro models of the human intestine[137]
-GlycerolMembranesCaffeine, ibuprofen, lidocaine and diclofenacTopical drug delivery systems[138]
--FilmCrocinTransdermal delivery of crocin[139]
PMGly—poly(N-methacryloyl glycine), MGly—N-methacryloylglycine, AAPH—2,2′-azobis(2-methylpropionamidine) dihydrochloride, MBA—N,N′-methylenebis(acrylamide).
Table 4. Diverse inorganic NC-based antimicrobial hybrids.
Table 4. Diverse inorganic NC-based antimicrobial hybrids.
Type of NCAntimicrobial Agent/
Compound
Hybrid FormSynthesis of HybridsMicrobial Growth InhibitionRef.
BNCAgNPsPorous hybridsImmobilization on the top and bottom surfaces of BNC by chemical interactionsE. coli, S. aureus[23]
CNFAgNPsCoated papersImpregnation of CNF with AgNPs’ solutionE. coli, S. aureus[156]
BNCAgNPsNanocompositesClassical Tollens reaction and reduction of AgNO3 with NaBH4S. aureus[157]
CNCZnOFilmsEvaporation-induced self-assembly in aqueous solutionE. coli, S. aureus[165]
TiO2
Ag2O
CNC/WGTiO2FilmsTiO2 NPs were added to the WG/CNC suspensionE. coli, S. cerevisiae, S. aureus[171]
CNFTiO2NanocompositesCross-linking of TiNPs with BTCA and SHPE. coli, S. aureus[172]
CNFCuOFreeze-dried nanofibrils“Green” reductive technique using an alcoholic extract of Terminalia chebula fruitC. albicans, E. coli, S. aureus[173]
CNCZnONanocompositesIn situ solution casting techniqueE. coli, S. aureus[174]
CNFZnOPaper sheetsElectrostatic assembly in aqueous medium using polyelectrolytes as macromolecular linkersB. cereus, K. pneumoniae, S. aureus[175]
BNCZnOFilmsPowdered BNC was dissolved in NMMO, and ZnO NPs were mixed into the BNC solution;E. coli[176]
BNCZnOSheetsUltrasonic-assisted in situ synthesisE. coli, S. aureus[177]
BNCMgOMembranesIn situ: Sonochemical and wet chemical methods
Ex situ: Immersion of BNC pellicles into a MgO dispersion
E. coli, S. aureus[178]
AgNPs—silver nanoparticles, WG—Wheat gluten, TiNPs—titanium nanoparticles, BTCA—1,2,3,4-butanetetracarboxylic acid, SHP—sodium hypophosphite, CuNPs—copper nanoparticles, NMMO—N-methylmorpholine-N-oxide.
Table 5. Diverse organic NC-based antimicrobial hybrids.
Table 5. Diverse organic NC-based antimicrobial hybrids.
Type of NCAntimicrobial Agent
(Additional Compound)
Hybrid FormSynthesis of HybridsMicrobial Growth InhibitionRef.
BNCBenzalkonium chlorideDry filmImmersion of BNC film into BZC solutionB. subtilis, S. aureus,
S. typhimurium
[187]
BNCPolyhexanide,
povidone-iodine
FleecesImmersion of BNC samples in PI or PHMBS. aureus[188]
CNF from ginger-FilmPreparation of film with chemicals (alkalization, bleaching, acid hydrolysis) and ultrasonicationB. subtilis, C. albicans,
E. coli, P. aeruginosa,
S. aureus
[194]
CNF from ginger-NanocompositesSolvent-casting method reinforcement using PS and TSB. cereus,
E. coli,
S. aureus,
S. typhimurium
[304]
CNF from ginger(Chitosan, PVA)NanocompositesSolvent-casting methodB. cereus,
E. coli,
S. aureus,
S. typhimurium
[305]
BNCChitosanHydrogelAdding CS in culture media during bacterial cultivationE. coli,
S. aureus
[220]
NCChitosanNanocompositesMixing of chitosan solution, NC solution and glycerol solutionE. coli,
S. aureus,
S. enteritidis
[306]
CNCChitosanFilmsFlax CNC incorporated in CS film solution by following the solution casting methodE. coli,
E. faecalis,
L. monocytogenes,
P. aeruginosa,
S. aureus
[307]
CNCChitosan
(PVP)
FilmSolution casting methodP. aeruginosa,
S. aureus
[222]
CNCChitosan, PCL,
grape seed extract
FilmCasting method for preparation of CS films with GSE and NC, the addition of PCL was achieved with coating and compression molding method for PCLE. coli,
L. monocytogenes
[234]
CNFChitosan, SNAPMembraneEncapsulation of SNAP in dispersed CS and mixed with CNFsE. faecalis,
L. monocytogenes,
S. aureus
[231]
BNCAPSMembraneSurface functionalization with aminoalkyl groupsE. coli,
S. aureus
[73]
BNCNisin, EDTAMembraneImmersion of BNC membranes into nisin solution with or without EDTAE. coli,
S. aureus
[243]
CNFNisinFilmImmobilization of nisin on CNF using the coupling agent (EDC-NHS)B. subtilis,
S. aureus
[308]
CNCNisin
(PLA)
FilmsPLA-CNC films were treated with nisin by adsorption/diffusion coating methodL. monocytogenes[309]
BNCBromelainMembraneSubmersion of BNC into a BL solutionE. coli,
P. aeruginosa,
S. aureus
[249]
BNCLaccaseMembranePhysical enzyme immobilization: immersion of BNC into a laccase preparationE. coli,
S. aureus
[254]
CNCCurcuminFilmCNF suspended in PVA solution, and then curcumin was addedB. coagulans,
C. albicans,
E. coli,
P. mirabilis,
S. aureus,
Streptococcus sp.
[108]
BNCFreeze-dried curcuminMembraneImmersion methodE. coli,
S. aureus
[310]
CNCCarvacrol, curcumin
(βCD, HPβCD)
FilmTOCNC-COONa and TOCNC-COOH were modified with βCD and HPβCD; Curcumin and carvacrol were entrapped by the attached HPβCDB. subtilis[311]
CNCRosinFilmEsterification on CNC using rosin as the grafting agent and reaction solventB. subtilis,
E. coli
[264]
CNFRosin,
(PLA/chitosan)
FilmCNF modified by rosin by the SolReact process and then used as a reinforcement filler within the PLA matrix; the film was further coated with CSB. subtilis,
E. coli
[312]
CNFLysozymeAerogelsPhysical immobilization of lysozyme: Mixing of the enzyme with CNFs followed by lyophilizationE. coli,
S. aureus
[267]
BNCLysozymeDispersionPhysical absorption method to immobilize lysozyme onto BC nanofibersA. niger,
E. coli,
L. monocytogenes,
S. aureus,
S. cerevisiae,
Y. enterocolitica
[313]
CNCAllicinNanocompositesModification of CNC with CA, further conjugation with allicin by a carbodiimide (EDC) cross-linker;A. niger, C. albicans,
E. coli,
S. aureus
[274]
LysozymeCoating of CNC with BSA and conjugated with lysozyme by the EDC method
NCAllicinFabricCellulose fabrics modified by APTES and conjugated with allicin-conjugated NC (EDC method)S. aureus[275]
CNCSavory essential oil
(agar)
FilmCNC suspension was dispersed in an AG solution; Tween 80 was added as the emulsifier, then SEO was added to the mixtureB. cereus,
E. coli,
L. monocytogenes,
S. aureus
[276]
BZC—benzalkonium chloride, PHMB—polyhexanide, PI—Povidone-iodine, PS—potato starch, TS—tapioca starch, PVA—poly(vinyl alcohol), CS—chitosan, PVP—poly(vinyl pyrrolidone), PCL—polycaprolactone, GSE—grape seed extract, SNAP—S-nitroso-N-acetyl-d-penicillamine, APS—3-aminopropyltrimethoxysilane, EDTA—ethylenediaminetetraacetic acid, EDC—1-ethyl-3-(3-dimethylaminopropyl)carbodiimide, NHS—N-hydroxysuccinimide, PLA—poly(lactic acid), BL—bromelain, βCD—beta-cyclodextrin, HPβCD—hydroxypropyl-beta-cyclodextrin, TOCNC-COONa—oxidized cellulose nanocrystals with sodium carboxylate groups, TOCN-COOH—oxidized cellulose nanocrystals with free carboxyl groups; CA—citric acid, BSA—bovine serum albumin, APTES—aminopropyl triethoxysilane, AG—agar, SEO—savory essential oil.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kupnik, K.; Primožič, M.; Kokol, V.; Leitgeb, M. Nanocellulose in Drug Delivery and Antimicrobially Active Materials. Polymers 2020, 12, 2825. https://doi.org/10.3390/polym12122825

AMA Style

Kupnik K, Primožič M, Kokol V, Leitgeb M. Nanocellulose in Drug Delivery and Antimicrobially Active Materials. Polymers. 2020; 12(12):2825. https://doi.org/10.3390/polym12122825

Chicago/Turabian Style

Kupnik, Kaja, Mateja Primožič, Vanja Kokol, and Maja Leitgeb. 2020. "Nanocellulose in Drug Delivery and Antimicrobially Active Materials" Polymers 12, no. 12: 2825. https://doi.org/10.3390/polym12122825

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