Open access peer-reviewed chapter

An Overview of Chitosan-Xanthan Gum Matrices as Controlled Release Drug Carriers

Written By

Suha M. Dadou, Milan D. Antonijevic, Babur Z. Chowdhry and Adnan A. Badwan

Submitted: 26 October 2017 Reviewed: 28 February 2018 Published: 18 July 2018

DOI: 10.5772/intechopen.76038

From the Edited Volume

Chitin-Chitosan - Myriad Functionalities in Science and Technology

Edited by Rajendra Sukhadeorao Dongre

Chapter metrics overview

1,600 Chapter Downloads

View Full Metrics

Abstract

Naturally occurring polysaccharides and/or their chemically modified derivatives have been widely investigated in relation to their use as components of controlled release systems for drug delivery. The aforementioned is due, in part, to their distinct properties such as abundant availability and biocompatibility as well as environmental and economic advantages. Chitosan (CS) and xanthan gum (XG) based matrices have received growing scientific/pharmaceutical interest as oral controlled release drug carriers. Herein, recent advances spanning the last two decades in CS-XG based drug delivery systems are reviewed with the emphasis being on oral tablet formulations, due to their versatility as pharmaceutical dosage forms. The mechanism of interaction between CS and XG, by means of computational and experimental approaches, is scrutinized. Results obtained from the literature establish the possibility of fabricating a controlled release drug delivery system based on CS and XG matrices. This can be achieved by monitoring and manipulating the physiochemical properties of the two polymers as well as the experimental variables affecting their drug retardation efficiency, without the need to employ special equipment or sophisticated experimental techniques/methodologies.

Keywords

  • drug delivery
  • controlled release
  • polymeric matrices
  • natural polysaccharides
  • xanthan gum
  • chitosan
  • polyelectrolyte complexes
  • molecular dynamics simulation

1. Introduction

The ultimate goal in drug design and development is to optimize a carrier that ensures the delivery of the active pharmaceutical ingredient(s) (APIs) to the systemic circulation in a safe and stable manner [1]. Patient compliance is a key aspect to consider when designing a new pharmaceutical dosage form [2]. Therefore, the way the drug will be introduced to the body should be optimized to ensure the availability of the drug at its site of action, at levels within the range of its therapeutic window (Figure 1).

Figure 1.

Plasma levels and therapeutic range of a drug following an oral administration of a single dose. MTC: minimum toxic concentration, and MEC: minimum effective concentration.

Despite emerging advances in drug delivery, the oral route remains the predominant route of drug administration. It is the simplest route, non-invasive and provides ~200 m2 of readily available surface area for drug absorption [3]. Conventional oral dosage forms usually release drugs immediately in the body, via first order release kinetics for both absorption and elimination processes [4]. Since the efficacy of the administered drug is limited to its residence time in plasma, frequent administration is required for APIs which exhibit a short biological half-life. As a result, low patient compliance and high fluctuation of drug levels in plasma is expected [5, 6]. In order to counter the foregoing drawbacks of conventional dosage forms, a new term in drug delivery was introduced; modified release dosage forms [7].

The United States Pharmacopeia defines modified release tablets as “coated or uncoated tablets that contain special excipients or are prepared by special procedures, or both, designed to modify the rate, the place or the time at which the active substance(s) are released”. Modified release delivery systems can be divided into delayed release systems and prolonged/extended release systems. Extended release delivery systems can further be subdivided into sustained and controlled release delivery systems, which differ in the rate at which they deliver APIs to the human blood circulation. Sustained release formulations function by continuously releasing APIs for a prolonged period of time. On the other hand, controlled release (CR) delivery systems do not only retard the release of the drug, but they deliver the drug to the body at a predetermined release rate or location [8]. Consequently, constant drug levels can be achieved (Figure 2).

Figure 2.

Comparison of plasma concentration-time profiles of drug release following multiple dosing from: (a) conventional, and (b) controlled-release dosage forms.

Advertisement

2. Controlled drug delivery

CR systems are composed of inactive pharmaceutical ingredient(s) that entrap the API(s) and release it/them at a time different from the immediate release form [4]. Researchers in the field of drug delivery have, and are currently still trying to acquire a better understanding of CR by attempting to integrate pharmaceutical technology with the relevant pharmacokinetic parameters associated with different drugs [9]. The rational underpinning controlled drug release includes, but are not limited to: masking the undesired side effects of drugs, attaining a constant drug release profile with minimal drug level fluctuations, and enhancing patient convenience by reducing administration frequency [10]. CR dosage forms are not only capable of extending the time over which drugs are released and providing constant drug levels but also with the potential of protecting therapeutic biomolecules such as peptides and proteins from enzymatic degradation in the gastrointestinal tract (GIT) [11]. CR systems can also be formulated to target the delivery of APIs to the desired site of action [12, 13].

Aside from the substantial need for CR formulations in drug delivery and the potential advantages they offer, the reproducibility and cost of equipment and techniques needed for the preparation of CR dosage forms on a large scale present a major obstacle towards the widespread production of CR delivery systems in pharmaceutical manufacturing. Figure 3 summarizes the key factors which require to be taken into account when optimizing a new CR dosage form.

Figure 3.

Key factors to be considered when developing a new dosage form in the pharmaceutical industry.

2.1. Design of CR systems

2.1.1. APIs

There are several criteria and properties that should be taken into consideration in the proposed use of an API when designing a controlled release formulation [14, 15].

  • The elimination half-life of the drug should be short. Drugs with long half-lives, greater than 8 h, provide a sustained release profile without the need to be formulated in a controlled release system.

  • Drugs with a wide therapeutic window are better candidates since higher doses need to be incorporated in CR formulations and dose dumping could occur.

  • The absorption rate of a candidate drug should be high to make sure that the release of drug from the CR delivery system is the rate determining step, not the absorption rate.

  • Drugs which exhibit high protein binding are retained in the plasma for a long time; thus, they do not require a CR delivery system.

  • Drugs that undergo extensive first pass metabolism are poor candidates for CR, since releasing the drug at lower rates will decrease its bioavailability. APIs with a bioavailability index higher than 75% are preferable.

2.1.2. Carriers and mechanism of drug release

Controlled drug release can be achieved by utilizing special techniques and devices. As the release of a drug from the delivery system is the rate limiting step in controlled release formulations, CR systems are classified according to the mechanism involved in drug release [3, 16]. In some preparations, more than one mechanism can be involved in the release of the API(s) from the CR systems (Figure 4).

Figure 4.

Classification of controlled release drug delivery systems combined with the main mechanisms involved in drug release and examples of inactive ingredients used to achieve CR.

2.1.3. In-vitro drug release kinetics

Since the objective of utilizing CR systems is to deliver a drug, or drugs, over a known time interval, several mathematical models (Table 1) have been suggested to describe drug release from the systems as a function of time.

ModelEquationMechanism of release
Zero order​​Q​ t​​ = ​Q​ 0​​ + ​K​ 0​​ t​Release is independent of drug concentration within the matrix or device
First order​Ln ​Q​ t​​ = Ln ​Q​ 0​​ + Kt​Release is dependent on drug concentration within the matrix or device
Higuchi​​Q​ t​​ = ​K​ H​​ ​t​​ 1/2​​Drug released via diffusion through an insoluble polymeric matrix
Hixon-Crowel​​​Q​ 0​​​​ 1/3​ − ​​Q​ t​​​​ 1/3​ = ​K​ HC​​ t​Drug release is dependent on drug dissolution rate in the media
Korsmeyer-Peppas​​Q​ t​​ / Q = K ​t​​ n​​This model is used when several mechanisms are involved in drug release from the system

Table 1.

Mathematical models of drug release kinetics from CR formulations*.

Where Q0 is the initial amount of drug in the dissolution media, Qt is the fraction released at time t, K is the rate release constant, and n is the release exponent.


Advertisement

3. Natural polysaccharides

Polymers are the most used materials to control the release of APIs. They can be classified as synthetic (silicons, polyesters and cellulose derivatives) and natural polymers (proteins and polysaccharides). Many naturally occurring polymers are inert, biodegradable, and cost-effective in relation to their industrial use [17]. In addition, their chemical structure can usually be easily modified to achieve the desired properties for a specific purpose [18]. Hence, the utilization of natural polymers as components of drug vehicles is gaining extensive attention [3]. The most used polymers are saccharides (carrageenan, cellulose) or proteins (collagen, gelatin) [19].

Natural polysaccharides are hydrophilic polymers consisting of repeating monosaccharide units linked via glycosidic bonds [20]. They are obtained from various sources, mainly vegetal (cellulose, starch), microbes (xanthan gum, dextran), crustaceans (chitin) and algae (alginate, carrageenan) [21, 22]. Depending on the identity of the constituent monomer(s), polysaccharides can be divided into homo-polysaccharides which are composed of the same repeating unit, such as cellulose, or hetero-polysaccharides which are built up from different saccharide units e.g., CS and XG [23, 24]. They can also be classified according to their ionic charge: non-polyelectrolyte (starch, cellulose), and polyelectrolyte polysaccharides. Polyelectrolytes are further sub-divided into negatively charged polymers; such as alginate and XG, or positively charged polymers, which are few in number, such as CS [25, 26].

The unique physicochemical characteristics of each polysaccharide are related to the type of monosaccharide building unit, position of the glycosidic bond, chain substitution and the overall molecular weight [25, 27]. Due to the presence of various functional groups attached to the polymer backbone (carboxyl ─COOH, amine ─NH2 and hydroxyl groups ─OH), polysaccharides have the ability to form non-covalent bonds with a wide range of synthetic and biological molecules [28, 29]. Moreover, they can attach to body tissues and mucus layers and sustain the release of encapsulated active ingredients [13]. The aforementioned properties have attracted attention towards the usage of polysaccharides in major industries including food, agronomy, cosmetics, biochemical engineering and pharmaceutical manufacturing [30, 31].

3.1. Chitosan (CS)

CS is a linear polysaccharide produced by the N-deacetylation of chitin [32]. Chitin is found mainly in the exoskeleton of marine crustaceans as well as insects and fungi [33]. Glucosamine and N-acetyl glucosamine are the building units of CS. They are linked via β(1-4)glycosidic bonds (Figure 5a). The degree of acetylation and distribution of acetyl groups along the polymer chain (either block or random distribution) are dependent on the duration of the deacetylation process and preparation method for CS [34, 35]. Following deacetylation of chitin, CS is (unlike chitin) soluble in acidic media. Moreover, the presence of primary amine groups leads to the unique properties of CS over all other natural polysaccharides [36]. It is the only saccharide possessing a high density positive net charge, which allows it to interact with a wide range of anionic polymers and biological molecules [32]. In addition, CS shows high mucoadhesion in the GIT which increases the residence time and enhances the permeation of active molecules [30]. Hence, CS is used commonly in the food industry, for pharmaceutical drug delivery and tissue engineering [37, 38].

Figure 5.

Schematic chemical structures of the building units of: (a) CS and (b) XG.

3.2. Xanthan gum (XG)

XG is a branched, hetero-polysaccharide produced via microbial fermentation of the microorganism Xanthomonas campestris [39]. The primary unit of XG (Figure 5b) consists of a cellulosic backbone composed of two d-glucose units (1-4) β-linked to a side-chain of d-mannose and d-glucuronic acid units at a ratio of 2:1, respectively [40]. d-Mannose, which is connected to the main backbone, is attached to an acetyl group at O6, while approximately half of the terminal d-mannose forms a pyruvic acid group between carbons C4 and C6. This side-chain is found at the O3 atom of each alternate glucose unit on the backbone. Due to the presence of carboxylic groups in its structure, XG exhibits a net negative charge and can form complexes with cationic polymers [41]. In the last decade, the demand for XG, in industry, has been increasing at about 5–10% per annum [42]. It is used in a broad variety of industries, including cosmetics, agriculture, food, textiles and oil [43, 44]. This is due to its safety (non-toxic), desirable rheological properties, high stability over a wide range of pH and temperature, together with its high resistance against enzymatic degradation [45, 46].

Advertisement

4. CS and XG matrices as controlled release drug delivery systems

4.1. Advances and applications

Matrix systems, based on polyelectrolyte polysaccharides, used to retard the release of APIs have been reported in the literature and some of them have been commercialized [29, 47]. The long term instability of their corresponding preparations due to the existence of charged groups limits their application in pharmaceutical manufacturing [48]. Introducing a cross-linker, such as tripolyphosphate or glutaraldehyde, to neutralize the polymeric matrix is a necessary approach to confront such a shortcoming. Though, substituting the cross-linker with an oppositely charged copolymer aids and abbets the synergistic effect of drug release retardation. XG proved to be a potential CR drug carrier. In aqueous solutions, XG shows high viscosity and water uptake capacity encapsulating the drug inside a thick gel-like layer which hinders the release of the incorporated drug. XG has been used alone and with other polymers such as HPMC, karaya gum, guar gum, and polyvinylpyrrolidone (PVP), or ethyl cellulose [49, 50, 51, 52, 53]. Formulated matrices were able to sustain the release of caffeine, azithromycin, ibuprofen and propranolol HCl. XG demonstrates a high capability of generating a near zero drug release profile.

Being the only known positively charged natural polymer in aqueous solutions, CS has been extensively investigated as a potential drug vehicle. CS has the ability to preserve the stability of active biomolecules, namely insulin, and enhance their absorption from the GIT [54, 55, 56]. CS was mixed with various polymers with the aim of modifying the release of active ingredients, protect genes and therapeutic peptides in the GIT and improve their permeation across the intestinal epithelium and to immobilize antibodies [57, 58, 59]. Alginate, carrageenan, pectin, hyaluronic acid and XG are amongst the many natural polymers to be used with CS [47, 60, 61].

The combination of CS and XG was first used in the form of a polyelectrolyte complex (PEC) hydrogel [62]. The hydrogels formed displayed pH dependent swelling behavior and addressed the possibility of developing a gastrointestinal drug delivery system. PECs are formed due to the attractive ionic forces between the positively charged amino groups in CS and the negatively charged carboxyl groups in XG [63]. Therefore, features of the PECs produced can be controlled by manipulating the physicochemical properties of each polymer [64]. Molecular weight, degree of acetylation (DA) of CS, and pyruvic acid content in XG are amongst the most crucial factors to be addressed [47, 65, 66]. Complexation conditions (including concentration of each polymer, mixing ratios, and pH) have a significant influence on the behavior and stability of the resulting PEC [67]. The combination of CS and XG has been extensively studied as a platform for CR drug delivery, resulting in many patents and the publication of research articles.

4.1.1. Patents on CS-XG based controlled release drug delivery system

CS-XG hydrogels have been studied in order to immobilize biological materials. This is due to their insolubility and high stability in acidic medium allowing the system to preserve biological activity and release the materials at neutral pH. CS-XG hydrogels served as a promising candidate for sustained release dosage forms [68]. CS-XG hydrogels were capable of stabilizing and controlling the release of highly sensitive active ingredients such as vitamins, amino acids, nucleic acids and polypeptides when applied topically or orally as dietary supplements [69]. Moreover, the prepared hydrogels were shown to play a role in regulating the dissolution rate of poorly water-soluble drugs as disclosed by the patent WO 2002003962 where fenofibrate, ursodeoxycholic acid, nifedipine and indomethacin were used as models of poorly water-soluble APIs [70].

Tablets comprising CS and XG as a hydrophilic matrix for oral controlled release were first presented by Badwan et al. [71]. A wide range of basic drugs were tested (e.g. ambroxol, salbutamol, metoclopramide, anti-infective, non-steroidal and anti-inflammatory agents (NSAIDs)). Tablets formulated using CS and XG have been used to deliver basic APIs in a controlled release pattern. The drug to polymer ratio used was 1:3, respectively and the preferred XG to CS ratio was 1:1. When the system was studied in-vivo on human volunteers, it produced constant serum levels of ambroxol over a period of 24 h. This study paved the way for further research on approaches and mechanisms involving tablet formulations based on the foregoing combination. A tablet dosage form based on CS-XG for the treatment of hypercholesterolemia was prepared [72]. A combination of lycopene and Monascus purpureus were used as active ingredients. When testing the preparation on human volunteers, a significant decrease in the plasma levels of cholesterol, LDL and triglycerides was reported. Moreover, HDL values were reported to be increased.

4.1.2. Research articles on CS-XG based controlled release drug delivery system

The applicability of CS-XG combinations to a wide range of dosage forms with different routes of administration has been investigated. Examples of the preparations reported in the literature together with a brief description of preparation methods, application and examples of incorporated APIs are summarized in Table 2.

Dosage formPreparation methodApplicationIncorporated ingredient
HydrogelsSolution mixing under heatDrug delivery, tissue engineering, immobilization of biological active materialsProbiotics [73], enzymes [64]
FilmsSolution castingDrug delivery, tissue engineering, food industryWound healing [74], amoxicillin [75], scaffolds [76]
CapsulesComplex coacervation, encapsulation of physically mixed powderDrug deliveryTheophylline [77], ciprofloxacin HCl [78]
BeadsExtrusion-dripping technique, complex coacervation mechanismDrug delivery, immobilization of biological active materialsProbiotics [79], glipizide [80], antibodies [59]
MicrospheresSpray drying, ionotropic gelation methodDrug deliveryMeclizine HCl [81]
Micro-emulsionsHomogenization with oil phaseDrug deliveryProgesterone [82]
Liposomes (chitosomes)Thin film hydration method, spray dryingDrug deliveryRifampicin [83]
CryogelFreeze-dryingImmobilization of biological active materialsEnzymes [84]
TabletsDirect compression, granulation, hot melt extrusionDrug deliveryMetformin HCl [85], terbutaline sulfate [48], propranolol [86]

Table 2.

Main applications of CS-XG based matrices.

4.2. Ionic interaction between XG and CS

In spite of the significant amount of research work conducted on XG and CS based matrices, a lack of understanding of the nature of the interaction between the two polymers and their behavior at the molecular level still exists. It was suggested that physico-chemical conditions in the stomach are an ideal environment for the formation of insoluble gels between the two polymers, which retards the release of APIs resulting in a sustained drug release profile [87]. Moreover, in vitro residence time evaluation on porcine mucin and in vivo studies using sheep models have addressed the bioadhesive nature of CS-XG matrices [88].

In order to acquire an understanding of the interaction between XG and CS and factors governing it, a molecular dynamics simulation (MDs) study was conducted by Dadou et al. [89]. The contribution of the DA of CS and protonation was evaluated. The resulting trajectories and binding free energy calculations revealed that electrostatic forces (polar interactions, ∆Eele) are the driving force for the interaction, and that the interaction occurs regardless of the DA and state of protonation of CS (free energy values are negative for all complexes). Protonation of CS molecules increases their penetration between the branched chains of XG and produces more stable complexes with lower free binding energy (Table 3). Intermolecular interactions (Van der Waals) showed a positive contribution to the formation of CS-XG PECs. This can be explained by the presence of a large number of hydroxyl groups along the chains of the polymers which can induce an instantaneous dipole attraction with the surrounding atoms. High positive solvation free energy (∆Gsolv) values justify the resultant insoluble PECs upon complexation between CS and XG in the laboratory. ∆Gsolv increases with protonation, reaching a maximum value when CS is fully protonated, indicating a higher extent of interaction with XG.

CS∆Eele∆EvdW∆Gsol∆G
0% P*, 0% DA−21.290−14.0321.890−13.43
50% P, 0% DA−227.53−24.47222.77−29.22
100% P, 0% DA−419.95−23.27412.57−30.65
0% P, 50% DA−25.080−21.1228.460−17.74
50% P, 50% DA−232.68−23.96227.36−29.28
0% P, 100% DA−25.070−25.7930.150−20.71

Table 3.

Binding free energy calculations for XG-CS complexes.

P represents state of protonation.


Values presented are in kcal/mol.

4.2.1. Mixing ratio

Since the interaction between CS and XG is electrostatically driven, the properties of the resultant PECs can be modified by controlling the net charge density. This can be achieved either by altering the mixing ratios or the initial concentrations of the polymeric solutions. Films of CS and XG were prepared and examined by scanning electron microscope (SEM) for additional information relating to the behavior and the interaction between the two polymers in aqueous solutions at different mixing ratios [90]. SEM images (Figure 6) show the rough surface of CS, whilst XG films produce a smooth surface. Combining the two polymers resulted in a pronounced alteration in the surface morphology of the films. The resulting PECs form irregular and fibrous surfaces with a porous structure. PECs at a mixing ratio of 1:1 (w/v %) showed a dramatic change in the surface structure and it is suggested that they represent the maximum interaction between the two polymers.

Figure 6.

SEM images at magnification power of x2000 of: (a) CS, (b) CS-XG (2:1), (c) CS-XG (1:1), (d) CS-XG (1:2) and (e) XG films, from Eftaiha et al. [90].

4.2.2. Initial concentration of XG

Argin-Soysal et al., studied the effect of polymer solution concentration on the formation of stable capsules and their subsequent swelling behavior [67]. The initial concentration of the XG solution was found to be the determining factor in relation to complexation density, more than CS. This is due its high molecular weight and the highly viscous hydrogels it forms when in contact with water [91]. The physical cross-linking between XG and CS was complete when the concentration of XG was 1.5%, regardless of other experimental conditions. Consequently, the degree of swelling was shown to be dictated by the initial aqueous concentration of XG.

4.2.3. pH and initial concentration of CS solutions

Dumitri et al., found that the pH of CS solutions has a moderate effect on the extent of interaction between XG and CS [65]. PECs where readily obtained within a wide range of pH (3.6–8.0). At lower pH values, the carboxyl groups of XG become protonated (uncharged) while the amine groups in CS are fully charged; hence, the interaction between CS and XG is impeded and reduced drug retardation occurs. The effect of pH was more pronounced when preparing low concentration solutions of CS. A considerable increase in the degree of swelling with the pH of solutions at CS concentrations of 0.65–0.7% (w/v) occurs.

4.2.4. Molecular weight (Mw) of CS

The swelling capacity of CS-XG based PECs were found to be influenced by the Mw of CS [68]. Lower water retention capacity was achieved by using a higher Mw of CS. The absorption of water increased noticeably with around 1000% weight gain at lower Mw of CS. The increase in water absorption causes the formation of more PEC layers which results in potentially more drug retardation. The aforementioned claim was supported by the slow release of diclofenac sodium from low molecular weight CS tablets (13 and 30 kDa) [92]. AlAkayleh et al., found that the release rate of terbutaline sulfate from XG-low molecular weight CS tablets (viscosity 38 mPa s) was slower than XG-high molecular weight CS (70 mPa s) [48].

4.2.5. DA of CS

The PEC between XG and CS is formed due to the electrostatic attraction between oppositely charged groups. Increasing the DA content decreases the number of available free amine groups that are readily protonated. In addition, the rigidity of CS chains increased with DA owing to strong intramolecular hydrogen bonds dictated by amide groups [93]. As a consequence, the extent of interaction between the polymers is reduced. Release of propranolol HCl from an CS-XG matrix was studied as a function of the degree of deacetylation (DDA) of CS [86]. Release of drug from the matrix was faster from the acetylated form of CS. This result is in accord with the outcomes of the molecular dynamics simulation study (Table 3) [89].

4.2.6. Ionic strength of solution

Adding ionic species to the solution resulted in a large decrease in water uptake of CS-XG PECs. Competition takes place between free ions and water molecules for the hydroxyl groups of the polymers and reduces the hydration of CS and XG chains. Thus, the degree of swelling is lower which, in turn, will have an effect on the drug retardation capability of the system [63].

4.2.7. Concentration of incorporated API

Hydrophilic matrices need to be used at high polymer to drug ratios in order to exert their effect in sustaining the release of APIs [27]. Thus, their application is restricted to low strength drugs, as addressed by Badwan et al. [71]. Al Remawi et al. studied the effect of polymer to drug ratio (P:D) on the release of ambroxol HCl from CS-XG based tablets [94]. The release rate of ambroxol was highly dependent on the P:D ratio. Greater retardation of drug release was attained at higher polymer ratios (Figure 7).

Figure 7.

In-vitro release of ambroxol HCl from: (R) reference product, prepared tablets at a P:D of (T1) 1:1, and (T2) 3:1, as reported by Al Remawi et al. [94].

Advertisement

5. Tablets comprising CS-XG

Oral solid dosage forms remain the most favorable choice to deliver APIs. The main reason is that they preserve the physicochemical stability of chemical entities more than liquid forms [95]. Additionally, tablets offer advantages for both manufacturers and patients which include ease of handling, low production cost, dose precision and self-administration capability [96].

Utilization of CS as an efficient excipient in tablet formulation is gradually increasing. CS powder exhibits a high surface area and porosity [93]. It produces tablets with high tensile strength that form a network-like structure when examined by microscopy [97]. The aim of using a combination of polymers, as tablet excipients, is to enhance compressibility and flowability properties. Furthermore, a polymeric mixture can increase the overall retardation performance of the system. CS-XG based tablets were formulated by compression of one layer and multi-layers; they were used solely or with other polymers such as galactomannan, seed gum or β-cyclodextrin [48, 85, 86, 98]. Moreover, they were used in immediate release, floating mucoadhesive and buccal tablets [99, 100]. According to Badwan et al., combining XG with CS has the advantage of improving the mechanical properties of both polymers [93].

5.1. Tablet preparation methods

5.1.1. Direct compression

Direct compression is a technique for formulating tablets which limits the use of solvents, temperature and equipment. It is the first choice whenever the API and inactive materials are suitable for direct compression and are stable at high pressure [101]. Powders of both active and inactive ingredients are mixed homogeneously, then sieved to the desired particle size. Finally, the prepared blend is compressed using a tablet press machine at a predetermined pressure [102]. CS-XG based tablets prepared via direct compression, showed a high potential towards sustaining the release of terbutaline sulfate and ambroxol [48, 103].

5.1.2. Dry granulation

Dry granulation is utilized to improve compaction properties of ingredients. It can influence flowability, stability, content uniformity of the powders and enhance the bioavailability of the API. This is attained by increasing the particle size of powder materials via aggregation of particles by either roller compaction or slugging and then milling to produce granules with the desired size [104].

5.1.3. Wet granulation

Wet granulation of tablet components is usually achieved using water, ethanol or a mixture of both. Following drying at an appropriate temperature, granules are mixed with other excipients if needed, passed through a sieve and finally compressed using a press machine at a predefined pressure [105]. Wet granulation is used to produce dust free granules, enhance flowability and cohesion. Eftaiha et al., investigated the ability of CS-XG tablets prepared by wet granulation using an aqueous solution of XG 1% (w/v) to modify the release of metronidazole [87]. The preparation was able to sustain the release of metronidazole, both in-vitro and in-vivo. A mucoadhesive behavior was observed when applying the tablets on sheep duodenum.

5.1.4. Hot melt extrusion (HME)

In HME the powdered API, functional polymers and any other excipients are blended in a mortar and pestle then fed into the hopper of a single or double screw extruder. Fukuda et al., prepared CS-XG tablets using HME to study the release of chlorpheniramine maleate [106]. The processing temperature was 90°C (zone 1), 95°C (zone 2), 105°C (zone 3) and 110°C (die) with a screw speed of 15 rpm. The processing time needed for powders inside the barrel of the extruder is usually ~3–4 min. The extruded materials were then manually cut into tablets of the desired weights. Chlorpheniramine release from the prepared CS-XG tablets occurred in a sustained manner and was independent of the pH and ionic strength of the dissolution media. HME offers the advantage of continuous processing and process analytical technology (PAT) which enables quality control testing throughout the process [107].

5.2. Mechanism of drug release

Drug release from CS-XG matrices is suggested to be governed by the dissolution rate of the drug and the polymers in the media as well as the diffusion of the drug from the matrices and erosion of the polymers. The data in Figure 8 illustrates the processes of drug release from a tablet composed of CS and XG. When the tablet is first exposed to aqueous media, an insoluble gel layer forms on the top surface of the tablet, as a result of polyelectrolyte complexation between the two charged polymers [108]. Water molecules start to penetrate this layer towards the matrix owing to the high water uptake capability of XG. [91]. Accordingly, both polymers and drug are dissolved and a rubbery hydrated region is formed (white area) [27]. On the other hand, a non-hydrated glassy area is formed at the core of the tablet, where no water molecules reach the system (black area) [109]. As time lapses, further penetration of water molecules into the tablet occurs resulting in the polymers chains being solvated. Consequently, swelling of the matrix occurs [110]. At this stage, water molecules enter between the polymer chains, the radius of gyration of the polymers increases and the end-to-end distance of the polymer backbones also increases [111]. This phenomenon of polymer relaxation is referred to as “swelling of the matrix” [91]. As more water molecules pass into the matrix, the polymer concentration on the outer surface of the tablet decreases, losing its integrity, and starts to dissolve in the medium. This phenomenon is termed “polymer erosion” [112].

Figure 8.

Schematic representation of the behavior of CS-XG tablets in an aqueous medium.

The rate of drug release from such a matrix could occur as a function of diffusion of the water molecules into the matrix, dissolution of both polymers and drug, polymer relaxation and erosion [91, 113]; this depends on the previously mentioned factors (Section 4.2) [111, 114].

Advertisement

6. Conclusions

Controlling the release of active ingredients is one of the fastest growing applications of CS-XG based matrices. Various drug delivery systems and newly emerging technologies have been developed in order to optimize the foregoing mixture. XG-CS matrices show a high potential towards controlling the release of a wide range of active biomolecules. The efficiency of CS-XG matrices to control the release of drugs can be reinforced by manipulating the physico-chemical properties of CS and XG and the experimental conditions used. Thus, incorporation/use of expensive devices and the method of preparation can be kept to a minimum. With further optimization and the utilization of newly emerging computational and quality by design tools, relatively simple and straightforward CS-XG based matrices can be formulated as potentially universal carriers to control the release of APIs.

Advertisement

Conflict of interest

The authors declare no competing financial interests.

References

  1. 1. Tibbitt MW, Dahlman JE, Langer R. Emerging frontiers in drug delivery. Journal of the American Chemical Society. 2016;138(3):704-717. DOI: 10.1021/jacs.5b09974
  2. 2. Bhowmik D, Gopinath H, Kumar BP, Duraivel S, Kumar KPS. Controlled release drug delivery systems. Pharma Innovation. 2012;1(10):24-32
  3. 3. Rosen H, Abribat T. The rise and rise of drug delivery. Nature Reviews. Drug Discovery. 2005;4(5):381-385. DOI: 10.1038/nrd1721
  4. 4. Perrie Y, Rades T. Chapter 1— Controlling drug delivery. In: Pharmaceutics—Drug Delivery and Targeting. 2nd ed. PhP; 2012. pp. 1-24
  5. 5. Kushal M, Monali M, Durgavati M, Mittal P, Umesh S, Pragna S. Oral controlled release drug delivery system: An overview. International Research Journal of Pharmacy. 2013;4(3):70-76. DOI: 10.7897/2230-8407.04312
  6. 6. Mrsny RJ. Oral drug delivery research in Europe. Journal of Controlled Release. 2012;161(2):247-253. DOI: 10.1016/j.jconrel.2012.01.017
  7. 7. Park K. Controlled drug delivery systems: Past forward and future back. Journal of Controlled Release. 2014;190:3-8. DOI: 10.1016/j.jconrel.2014.03.054
  8. 8. Crommelin DJA, Florence AT. Towards more effective advanced drug delivery systems. International Journal of Pharmaceutics. 2013;454(1):496-511. DOI: 10.1016/j.ijpharm.2013.02.020
  9. 9. Hoffman AS. The origins and evolution of “controlled” drug delivery systems. Journal of Controlled Release. 2008;132(3):153-163. DOI: 10.1016/j.jconrel.2008.08.012
  10. 10. Lee PI, Li JX. Evolution of oral controlled release dosage forms. In: Wen H, Park K, editors. Oral Controlled Release Formulation Design and Drug Delivery: Theory to Practice. Hoboken, NJ: John Wiley & Sons, Inc.; 2010. pp. 21-31. DOI: 10.1002/9780470640487.ch2
  11. 11. Patel G, Misra A. Oral delivery of proteins and peptides: Concepts and applications. In: Ambikanandan M, editors. Challenges in Delivery of Therapeutic Genomics and Proteomics. London: Elsevier; 2011. pp. 481-529. DOI: 10.1016/B978-0-12-384964-9.00010-4
  12. 12. Siegel RA, Rathbone MJ. Overview of controlled release mechanisms. In: Siepmann J, Siegel RA, Rathbone MJ, editors. Fundamentals and Applications of Controlled Release Drug Delivery. New York: Springer; 2012. pp. 19-43. DOI: 10.1007/978-1-4614-0881-9_2
  13. 13. Sinha VR, Kumria R. Polysaccharides in colon-specific drug delivery. International Journal of Pharmaceutics. 2001;224(1-2):19-38. DOI: 10.1016/S0378-5173(01)00720-7
  14. 14. Chrzanowski F. Preformulation considerations for controlled release dosage forms: Part I selecting candidates. AAPS PharmSciTech. 2008;9(2):635-638. DOI: 10.1208/s12249-008-9068-2
  15. 15. Mahler S, Roy I. Advances in drug delivery. Journal of Chemical Technology and Biotechnology. 2015;90(7):1167-1168. DOI: 10.1002/jctb.4689
  16. 16. Holowka EP, Bhatia SK. Controlled-release systems. In: Drug Delivery: Materials Design and Clinical Perspective. New York: Springer; 2014. pp. 7-62. DOI: 10.1007/978-1-4939-1998-7
  17. 17. Hovgaard L, Brondsted H. Current applications of polysaccharides in colon targeting. Critical Reviews in Therapeutic Drug Carrier Systems. 1996;13(3-4):185-223
  18. 18. Posocco B, Dreussi E, De Santa J, et al. Polysaccharides for the delivery of antitumor drugs. Materials (Basel). 2015;8(5):2569-2615. DOI: 10.3390/ma8052569
  19. 19. Malafaya PB, Silva GA, Reis RL. Natural-origin polymers as carriers and scaffolds for biomolecules and cell delivery in tissue engineering applications. Advanced Drug Delivery Reviews. 2007;59(4-5):207-233. DOI: 10.1016/j.addr.2007.03.012
  20. 20. Zhang N, Wardwell PR, Bader RA. Polysaccharide-based micelles for drug delivery. Pharmaceutics. 2013;5(2):329-352. DOI: 10.3390/pharmaceutics5020329
  21. 21. Raemdonck K, Martens TF, Braeckmans K, Demeester J, De Smedt SC. Polysaccharide-based nucleic acid nanoformulations. Advanced Drug Delivery Reviews. 2013;65(9):1123-1147. DOI: 10.1016/j.addr.2013.05.002
  22. 22. Yang J, Han S, Zheng H, Dong H, Liu J. Preparation and application of micro/nanoparticles based on natural polysaccharides. Carbohydrate Polymers. 2015;123:53-66. DOI: 10.1016/j.carbpol.2015.01.029
  23. 23. Debele TA, Mekuria SL, Tsai HC. Polysaccharide based nanogels in the drug delivery system: Application as the carrier of pharmaceutical agents. Materials Science and Engineering: C. 2016;68:964-981. DOI: 10.1016/j.msec.2016.05.121
  24. 24. Gulrez SKH, Al-Assaf S, Phillips GO. Hydrogels: Methods of preparation, characterisation and applications. In: Carpi A, editors. Progress in Molecular and Environmental Bioengineering—From Analysis and Modeling to Technology Applications. Rijeka, Croatia: InTech; 2011. pp. 117-150. DOI: 10.5772/24553
  25. 25. Liu Z, Jiao Y, Wang Y, Zhou C, Zhang Z. Polysaccharides-based nanoparticles as drug delivery systems. Advanced Drug Delivery Reviews. 2008;60(15):1650-1662. DOI: 10.1016/j.addr.2008.09.001
  26. 26. Alvarez-Lorenzo C, Blanco-Fernandez B, Puga AM, Concheiro A. Crosslinked ionic polysaccharides for stimuli-sensitive drug delivery. Advanced Drug Delivery Reviews. 2013;65(9):1148-1171. DOI: 10.1016/j.addr.2013.04.016
  27. 27. Maderuelo C, Zarzuelo A, Lanao JM. Critical factors in the release of drugs from sustained release hydrophilic matrices. Journal of Controlled Release. 2011;154(1):2-19. DOI: 10.1016/j.jconrel.2011.04.002
  28. 28. Rinaudo M. Non-covalent interactions in polysaccharide systems. Macromolecular Bioscience. 2006;6(8):590-610. DOI: 10.1002/mabi.200600053
  29. 29. Shelke NB, James R, Laurencin CT, Kumbar SG. Polysaccharide biomaterials for drug delivery and regenerative engineering. Polymers for Advanced Technologies. 2014;25(5):448-460. DOI: 10.1002/pat.3266
  30. 30. Klein S. Polysaccharides in oral drug delivery—Recent applications and future perspectives. In: Polysaccharide Materials: Performance by Design. Vol. 1017. ACS Symposium Series. American Chemical Society; 2009. pp. 1-13. DOI: 10.1021/bk-2009-1017.ch001
  31. 31. Goodarzi N, Varshochian R, Kamalinia G, Atyabi F, Dinarvand R. A review of polysaccharide cytotoxic drug conjugates for cancer therapy. Carbohydrate Polymers. 2013;92(2):1280-1293. DOI: 10.1016/j.carbpol.2012.10.036
  32. 32. Kushwaha Swatantra KS, Rai Awani K, Satyawan S. Chitosan: A platform for targeted drug delivery. International Journal PharmTech, Research. 2010;2(4):2271-2282
  33. 33. Boonsongrit Y, Mitrevej A, Mueller BW. Chitosan drug binding by ionic interaction. European Journal of Pharmaceutics and Biopharmaceutics. 2006;62(3):267-274. DOI: 10.1016/j.ejpb.2005.09.002
  34. 34. Lu B, Wang C-F, Wu D-Q, Li C, Zhang X-Z, Zhuo R-X. Chitosan based oligoamine polymers: Synthesis, characterization, and gene delivery. Journal of Controlled Release. 2009;137(1):54-62. DOI: 10.1016/j.jconrel.2009.03.004
  35. 35. Chang KLB, Tsai G, Lee J, Fu WR. Heterogeneous N-deacetylation of chitin in alkaline solution. Carbohydrate Research. 1997;303(3):327-332. DOI: 10.1016/S0008-6215(97)00179-1
  36. 36. Brugnerotto J, Desbrieres J, Heux L, Mazeau K, Rinaudo M. Overview on structural characterization of chitosan molecules in relation with their behavior in solution. In: Macromolecular Symposia. WILEY‐VCH Verlag GmbH; 2001;168(1):1-20. DOI: 10.1002/1521-3900(200103)168:1<1::AID-MASY1>3.0.CO;2-W
  37. 37. Amidi M, Mastrobattista E, Jiskoot W, Hennink WE. Chitosan-based delivery systems for protein therapeutics and antigens. Advanced Drug Delivery Reviews. 2010;62(1):59-82. DOI: 10.1016/j.addr.2009.11.009
  38. 38. Qinna NA, Akayleh FT, Al Remawi MM, Kamona BS, Taha H, Badwan AA. Evaluation of a functional food preparation based on chitosan as a meal replacement diet. Journal of Functional Foods. 2013;5(3):1125-1134. DOI: 10.1016/j.jff.2013.03.009
  39. 39. Christensen BE, Smidsrød O. Hydrolysis of xanthan in dilute acid: Effects on chemical composition, conformation, and intrinsic viscosity. Carbohydrate Research. 1991;214(1):55-69. DOI: 10.1016/S0008-6215(00)90530-5
  40. 40. Arendt O, Kulicke W-M. Determination of the viscoelastic properties of a homologous series of the fermentation polymer xanthan gum. Die Angewandte Makromolekulare Chemie. 1998;259(1):61-67. DOI: 10.1002/(SICI)1522-9505(19981001)259:1<61::AID-APMC61>3.0.CO;2-V
  41. 41. García-Ochoa F, Santos VE, Casas JA, Gómez E. Xanthan gum: Production, recovery, and properties. Biotechnology Advances. 2000;18(7):549-579. DOI: 10.1016/S0734-9750(00)00050-1
  42. 42. Lo YM, Yang ST, Min DB. Effects of yeast extract and glucose on xanthan production and cell growth in batch culture of Xanthomonas campestris. Applied Microbiology and Biotechnology. 1997;47(6):689-694. DOI: 10.1007/s002530050996
  43. 43. Benny IS, Gunasekar V, Ponnusami V. Review on application of xanthan gum in drug delivery. International Journal PharmTech, Research. 2014;6(4):1322-1326
  44. 44. Faria S, De Oliveira Petkowicz CL, De Morais SAL, et al. Characterization of xanthan gum produced from sugar cane broth. Carbohydrate Polymers. 2011;86(2):469-476. DOI: 10.1016/j.carbpol.2011.04.063
  45. 45. Bueno VB, Petri DFS. Xanthan hydrogel films: Molecular conformation, charge density and protein carriers. Carbohydrate Polymers. 2014;101(1):897-904. DOI: 10.1016/j.carbpol.2013.10.039
  46. 46. Leela JK, Sharma G. Studies on xanthan production from Xanthomonas campestris. Bioprocess Engineering. 2000;23(6):687-689. DOI: 10.1007/s004499900054
  47. 47. Luo Y, Wang Q. Recent development of chitosan-based polyelectrolyte complexes with natural polysaccharides for drug delivery. International Journal of Biological Macromolecules. 2014;64:353-367. DOI: 10.1016/j.ijbiomac.2013.12.017
  48. 48. Al-Akayleh F, Al Remawi M, Rashid I, Badwan A. Formulation and In vitro assessment of sustained release terbutaline sulfate tablet made from binary hydrophilic polymer mixtures. Pharmaceutical Development and Technology. 2013;18(5):1204-1212. DOI: 10.3109/10837450.2011.620968
  49. 49. Munday DL, Cox PJ. Compressed xanthan and karaya gum matrices: Hydration, erosion and drug release mechanisms. International Journal of Pharmaceutics. 2000;203(1-2):179-192. DOI: 10.1016/S0378-5173(00)00444-0
  50. 50. Santos H, Veiga F, Pina ME, Sousa JJ. Compaction, compression and drug release properties of diclofenac sodium and ibuprofen pellets comprising xanthan gum as a sustained release agent. International Journal of Pharmaceutics. 2005;295(1-2):15-27. DOI: 10.1016/j.ijpharm.2005.01.014
  51. 51. Mughal MA, Iqbal Z, Neau SH. Guar gum, xanthan gum, and HPMC can define release mechanisms and sustain release of propranolol hydrochloride. AAPS PharmSciTech. 2011;12(1):77-87. DOI: 10.1208/s12249-010-9570-1
  52. 52. Talukdar MM, Plaizier-Vercammen J. Evaluation of xanthan cum as a hydrophilic matrix for controlled-release dosage form preparations. Drug Development and Industrial Pharmacy. 1993;19(9):1037-1046. DOI: 10.3109/03639049309062999
  53. 53. Verhoeven E, Vervaet C, Remon JP. Xanthan gum to tailor drug release of sustained-release ethylcellulose mini-matrices prepared via hot-melt extrusion: In vitro and in vivo evaluation. European Journal of Pharmaceutics and Biopharmaceutics. 2006;63(3):320-330. DOI: 10.1016/j.ejpb.2005.12.004
  54. 54. Elsayed A, Al-Remawi M, Qinna N, Farouk A, Al-Sou’od KA, Badwan AA. Chitosan–sodium lauryl sulfate nanoparticles as a carrier system for the in vivo delivery of oral insulin. AAPS PharmSciTech. 2011;12(3):958-964. DOI: 10.1208/s12249-011-9647-5
  55. 55. Rekha MR, Sharma CP. Synthesis and evaluation of lauryl succinyl chitosan particles towards oral insulin delivery and absorption. Journal of Controlled Release. 2009;135(2):144-151. DOI: 10.1016/j.jconrel.2009.01.011
  56. 56. Elsayed A, Remawi MA, Qinna N, Farouk A, Badwan A. Formulation and characterization of an oily-based system for oral delivery of insulin. European Journal of Pharmaceutics and Biopharmaceutics. 2009;73(2):269-279. DOI: 10.1016/j.ejpb.2009.06.004
  57. 57. Bhattarai N, Gunn J, Zhang M. Chitosan-based hydrogels for controlled, localized drug delivery. Advanced Drug Delivery Reviews. 2010;62(1):83-99. DOI: 10.1016/j.addr.2009.07.019
  58. 58. Chen MC, Mi FL, Liao ZX, et al. Recent advances in chitosan-based nanoparticles for oral delivery of macromolecules. Advanced Drug Delivery Reviews. 2013;65(6):865-879. DOI: 10.1016/j.addr.2012.10.010
  59. 59. Albarghouthi M, Fara DA, Saleem M, El-Thaher T, Matalka K, Badwan A. Immobilization of antibodies on alginate-chitosan beads. International Journal of Pharmaceutics. 2000;206(1-2):23-34. DOI: 10.1016/S0378-5173(00)00470-1
  60. 60. Li L, Wang L, Shao Y, Ni R, Zhang T, Mao S. Drug release characteristics from chitosan-alginate matrix tablets based on the theory of self-assembled film. International Journal of Pharmaceutics. 2013;450(1-2):197-207. DOI: 10.1016/j.ijpharm.2013.04.052
  61. 61. Volod’Ko AV, Davydova VN, Chusovitin E, et al. Soluble chitosan-carrageenan polyelectrolyte complexes and their gastroprotective activity. Carbohydrate Polymers. 2014;101(1):1087-1093. DOI: 10.1016/j.carbpol.2013.10.049
  62. 62. Chu CH, Sakiyama T, Yano T. Ph-sensitive swelling of a polyelectrolyte complex gel prepared from xanthan and chitosan. Bioscience, Biotechnology, and Biochemistry. 1995;59(4):717-719. DOI: 10.1080/bbb.59.717
  63. 63. Martínez-Ruvalcaba A, Chornet E, Rodrigue D. Viscoelastic properties of dispersed chitosan/xanthan hydrogels. Carbohydrate Polymers. 2007;67(4):586-595. DOI: 10.1016/j.carbpol.2006.06.033
  64. 64. Dumitriu S, Chornet E. Immobilization of xylanase in chitosan-xanthan hydrogels. Biotechnology Progress. 1997;13:539-545. DOI: 10.1021/bp970059i
  65. 65. Magnin D, Dumitriu S, Chornet E. Immobilization of enzymes into a polyionic hydrogel: ChitoXan. Journal of Bioactive and Compatible Polymers. 2003;18(5):355-373. DOI: 10.1177/088391103038375
  66. 66. Qinna AN, Karwi GQ, Al-Jbour N, et al. Influence of molecular weight and degree of deacetylation of low molecular weight chitosan on the bioactivity of oral insulin preparations. Marine Drugs. 2015;13(4):1710-1725. DOI: 10.3390/md13041710
  67. 67. Argin-Soysal S, Kofinas P, Lo YM. Effect of complexation conditions on xanthan–chitosan polyelectrolyte complex gels. Food Hydrocolloids. 2009;23(1):202-209. DOI: https://doi.org/10.1016/j.foodhyd.2007.12.011
  68. 68. Dumitriu S, Chornet E, Vidal P. Polyionic Insoluble Hydrogels Comprising Xanthan and Chitosan. U.S. Patent No. 5,620,706. Washington, DC. April 15, 1997. http://www.google.com/patents/US5620706
  69. 69. Chornet E, Dumitriu S. Polyionic hydrogels based on xanthan and chitosan for stabilising and controlled release of vitamins. January 2000. http://www.google.com/patents/WO2000004086A1?cl=en
  70. 70. Chornet E, Ishizawa C, Dumitriu S. Drug delivery system for poorly water soluble drugs. January 2002. https://www.google.ch/patents/WO2002003962A2?cl=en
  71. 71. Badwan AA, Al-Remawi M, Salem M. Universal controlled-release composition comprising chitosan. Eur Pat EP1512394. 2008
  72. 72. Mangiapane A. Combination of substances for the treatment of patients with hypercholesterolemia and related disorders. May 2012. https://www.google.ch/patents/EP2455073A1?cl=en
  73. 73. Chu C-H, Kumagai H, Nakamura K. Application of polyelectrolyte complex gel composed of xanthan and chitosan to the immobilization of corynebacterium glutamicum. Journal of Applied Polymer Science. 1996;60(7):1041-1047. DOI: 10.1002/(SICI)1097-4628(19960516)60:7<1041::AID-APP15>3.0.CO;2-3
  74. 74. Bellini MZ, Caliari-Oliveira C, Mizukami A, et al. Combining xanthan and chitosan membranes to multipotent mesenchymal stromal cells as bioactive dressings for dermo-epidermal wounds. Journal of Biomaterials Applications. 2014;29(8):1155-1166. DOI: 10.1177/0885328214553959
  75. 75. Thakur A, Monga S, Wanchoo RK. Sorption and drug delease studies from semi-interpenetrating polymer networks of chitosan and xanthan gum. Chemical and Biochemical Engineering Quarterly. 2014;28(1):105-115
  76. 76. Veiga IG, Moraes ÂM. Study of the swelling and stability properties of chitosan-xanthan membranes. Journal of Applied Polymer Science. 2012;124(Suppl. 1):E154-E160. DOI: 10.1002/app.35526
  77. 77. Popa N, Novac O, Profire L, Lupusoru CE, Popa MI. Hydrogels based on chitosan-xanthan for controlled release of theophylline. Journal of Materials Science. Materials in Medicine. 2010;21(4):1241-1248. DOI: 10.1007/s10856-009-3937-4
  78. 78. Verma A, Bansal A, Ghosh A, Pandit J. Low molecular mass chitosan as carrier for a hydrodynamically balanced system for sustained delivery of ciprofloxacin hydrochloride. Acta Pharmaceutica. 2012;62(2):237-250. DOI: 10.2478/v10007-012-0013-2
  79. 79. Fareez IM, Lim SM, Mishra RK, Ramasamy K. Chitosan coated alginate-xanthan gum bead enhanced pH and thermotolerance of Lactobacillus plantarum LAB12. International Journal of Biological Macromolecules. 2015;72:1419-1428. DOI: 10.1016/j.ijbiomac.2014.10.054
  80. 80. Kulkarni N, Wakte P, Naik J. Development of floating chitosan-xanthan beads for oral controlled release of glipizide. International Journal of Pharmaceutical Investigation. 2015;5(2):73-80. DOI: 10.4103/2230-973X.153381
  81. 81. Kharshoum RM, Aboutaleb HA. Formulation, development and evaluation of meclozine hydrochloride microspheres. Journal of Bioequivalence and Bioavailability. 2016;8(1):27-32. DOI: 10.4172/jbb.1000262
  82. 82. Cornas F, Dolz M, Herraez M, Diez-Sales O. Rheological properties of progesterone microemulsions: Influence of xanthan and chitosan biopolymer concentration. Journal of Applied Polymer Science. 2008;110(2):1225-1235. DOI: 10.1002/app.28657
  83. 83. Manca ML, Manconi M, Valenti D, et al. Liposomes coated with chitosan-xanthan gum (chitosomes) as potential carriers for pulmonary delivery of rifampicin. Journal of Pharmaceutical Sciences. 2012;101(2):566-575. DOI: 10.1002/jps.22775
  84. 84. Liu H, Nakagawa K, Kato DI, Chaudhary D, Tadé MO. Enzyme encapsulation in freeze-dried bionanocomposites prepared from chitosan and xanthan gum blend. Materials Chemistry and Physics. 2011;129(1-2):488-494. DOI: 10.1016/j.matchemphys.2011.04.043
  85. 85. Corti G, Cirri M, Maestrelli F, Mennini N, Mura P. Sustained-release matrix tablets of metformin hydrochloride in combination with triacetyl-β-cyclodextrin. European Journal of Pharmaceutics and Biopharmaceutics. 2008;68(2):303-309. DOI: 10.1016/j.ejpb.2007.06.004
  86. 86. Phaechamud T, Ritthidej GC. Sustained-release from layered matrix system comprising chitosan and xanthan gum. Drug Development and Industrial Pharmacy. 2007;33(6):595-605. DOI: 10.1080/03639040601015521
  87. 87. Eftaiha AF, Qinna N, Rashid IS, et al. Bioadhesive controlled metronidazole release matrix based on chitosan and xanthan gum. Marine Drugs. 2010;8(5):1716-1730. DOI: 10.3390/md8051716
  88. 88. Mura P, Cirri M, Mennini N, Casella G, Maestrelli F. Polymeric mucoadhesive tablets for topical or systemic buccal delivery of clonazepam: Effect of cyclodextrin complexation. Carbohydrate Polymers. 2016;152:755-763. DOI: 10.1016/j.carbpol.2016.07.075
  89. 89. Dadou SM, El-Barghouthi MI, Alabdallah SK, Badwan AA, Antonijevic MD, Chowdhry BZ. Effect of protonation state and N-acetylation of chitosan on its interaction with xanthan gum: A molecular dynamics simulation study. Marine Drugs. 2017;15(10):298. DOI: 10.3390/md15100298
  90. 90. Eftaiha AF, El-Barghouthi MI, Rashid IS, Al-Remawi MM, Saleh AI, Badwan AA. Compressibility and compactibility studies of chitosan, xanthan gum, and their mixtures. Journal of Materials Science. 2009;44(4):1054-1062. DOI: 10.1007/s10853-008-3186-9
  91. 91. Ghori MU, Conway BR. Hydrophilic matrices for oral control drug delivery. American Journal of Pharmacology and Toxicology. 2015;3(5):103-109. DOI: 10.12691/AJPS-3-5-1
  92. 92. Alakayleh F, Rashid I, Al-Omari MMH, Al-Sou’od K, Chowdhry BZ, Badwan AA. Compression profiles of different molecular weight chitosans. Powder Technology. 2016;299:107-118. DOI: 10.1016/j.powtec.2016.05.019
  93. 93. Badwan AA, Rashid I, Al Omari MMH, Darras FH. Chitin and chitosan as direct compression excipients in pharmaceutical applications. Marine Drugs. 2015;13(3):1519-1547. DOI: 10.3390/md13031519
  94. 94. Al Remawi M, Al-Akayleh F, Salem MS, Al Shami M, Badwan A. Application of an excipient made from chitosan and xanthan gum as a single component for the controlled release of Ambroxol. Journal of Excipients and Food Chemicals. 2013;4(2):48-57
  95. 95. Gad SC, editors. Pharmaceutical Manufacturing Handbook: Production and Processes. New Jersey: John Wiley & Sons, Inc.; 2007. DOI: 10.1002/9780470259818
  96. 96. Patel JS. A review on bilayer tablets. Journal of Drug Discovery and Therapeutics. 2013;1(3):40-48
  97. 97. Picker-Freyer KM, Brink D. Evaluation of powder and tableting properties of chitosan. AAPS PharmSciTech. 2006;7(3):75. DOI: 10.1208/pt070375
  98. 98. Nogueira CC dos S, Cabral LM, Santos TC dos, Marucci A, Alhaique F. Evaluation of new polysaccharides networks for extended-release purposes: Mesquite seed gum (MSG), xanthan gum and chitosan. Revista Brasileira de Ciências Farmacêuticas. 2003;39:273-288. DOI: 10.1590/S1516-93322003000300007
  99. 99. Azhar SA, Rajesh Kumar P, Vivek S, Somashekar S. Studies on directly compressed ondansetron hydrochloride mucoadhesive buccal tablets using gelatin, chitosan and xanthan gum along with HPMC K4M. Journal of Applied Pharmaceutical Science. 2012;2(5):100-105. DOI: 10.7324/JAPS.2012.2517
  100. 100. Panigrahy RN, Mahale AM, Dhaked PS. Formulation and in vitro evaluation of combined floating-mucoadesive tablet of Metoprolol succinate. International Journal of Pharmacy and Pharmaceutical Sciences. 2011;3(2):221-226
  101. 101. Bushra R, Shoaib MH, Aslam N, Hashmat D. Formulation development and optimization of ibuprofen tablets by direct compression method. Pakistan Journal of Pharmaceutical Sciences. 2008;21(2):113-120. DOI: 10.1002/adv.21536
  102. 102. Al-Remawi MMA, Badwan AA, Salem MBAWS. Universal controlled-release composition comprising chitosan. March 2005. http://www.google.com/patents/EP1512394A1?cl=en
  103. 103. Al-Akayleh F, Al Remawi M, Salem MS, Badwan A. Using chitosan and xanthan gum mixtures as excipients in controlled release formulations of ambroxol HCl—In vitro drug release and swelling behavior. Journal of Excipients and Food Chemicals. 2014;5(2):140-148
  104. 104. Kleinebudde P. Roll compaction/dry granulation: Pharmaceutical applications. European Journal of Pharmaceutics and Biopharmaceutics. 2004;58(2):317-326. DOI: 10.1016/j.ejpb.2004.04.014
  105. 105. Shao Y, Li L, Gu X, Wang L, Mao S. Evaluation of chitosan-anionic polymers based tablets for extended-release of highly water-soluble drugs. Asian Journal of Pharmaceutical Sciences. 2015;10(1):24-30. DOI: 10.1016/j.ajps.2014.08.002
  106. 106. Fukuda M, Peppas NA, McGinity JW. Properties of sustained release hot-melt extruded tablets containing chitosan and xanthan gum. International Journal of Pharmaceutics. 2006;310(1-2):90-100. DOI: 10.1016/j.ijpharm.2005.11.042
  107. 107. Patil H, Tiwari RV, Repka MA. Hot-melt extrusion: From theory to application in pharmaceutical formulation. AAPS PharmSciTech. 2016;17(1):20-42. DOI: 10.1208/s12249-015-0360-7
  108. 108. Tiwari SB, Rajabi-Siahboomi AR. Modulation of drug release from hydrophilic matrices. PharmTech Europe. 2008;20(9):24-32
  109. 109. Nokhodchi A, Raja S, Patel P, Asare-Addo K. The role of oral controlled release matrix tablets in drug delivery systems. BioImpacts: BI. 2012;2(4):175-187. DOI: 10.5681/bi.2012.027
  110. 110. Rohindra D, Nand A, Khurma J. Swelling properties of chitosan hydrogels. South Pacific Journal of Natural and Applied Sciences. 2004;22(1):32-35. DOI: 10.1071/SP04005
  111. 111. Huanbutta K, Sriamornsak P, Limmatvapirat S, et al. Swelling kinetics of spray-dried chitosan acetate assessed by magnetic resonance imaging and their relation to drug release kinetics of chitosan matrix tablets. European Journal of Pharmaceutics and Biopharmaceutics. 2011;77(2):320-326. DOI: 10.1016/j.ejpb.2010.11.019
  112. 112. Ferrero C, Muñoz-Ruiz A, Jiménez-Castellanos MR. Fronts movement as a useful tool for hydrophilic matrix release mechanism elucidation. International Journal of Pharmaceutics. 2000;202(1-2):21-28. DOI: 10.1016/S0378-5173(00)00407-5
  113. 113. Kim H, Fassihi R. Application of binary polymer system in drug release rate modulation. 2. Influence of formulation variables and hydrodynamic conditions on release kinetics. Journal of Pharmaceutical Sciences. 1997;86(3):323-328. DOI: 10.1021/js960307p
  114. 114. Mitchell K, Ford JL, Armstrong DJ, Elliott PNC, Rostron C, Hogan JE. The influence of concentration on the release of drugs from gels and matrices containing Methocel®. International Journal of Pharmaceutics. 1993;100(1-3):155-163. DOI: 10.1016/0378-5173(93)90086-U

Written By

Suha M. Dadou, Milan D. Antonijevic, Babur Z. Chowdhry and Adnan A. Badwan

Submitted: 26 October 2017 Reviewed: 28 February 2018 Published: 18 July 2018