iranian journal chitosan

Upload: eva-pae-o

Post on 14-Apr-2018

222 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/29/2019 Iranian Journal Chitosan

    1/12

    Iranian Polymer Journal

    20 (5), 2011, 445-456

    alginate;

    drug delivery system;burst release;

    ionic gelation;

    tripolyphosphate.

    (*) To whom correspondence to be addressed.

    E-mail: [email protected]

    A B S T R A C T

    Key Words:

    Reinforcement of Chitosan Nanoparticles Obtained

    by an Ionic Cross-linking Process

    Morteza Hasanzadeh Kafshgari, Mohammad Khorram*, Mobina Khodadoost,

    and Sahar Khavari

    Department of Chemical Engineering, University of Sistan and Baluchestan,

    P.O. Box: 98161/164, Zahedan, Iran

    Received 22 February 2010; accepted 27 November 2010

    The reduction in burst release of a new drug nanocarrier system was achieved by

    ionic gelation of nanoparticles made of chitosan and tripolyphosphate. Capability

    of calcium alginate for reinforcement of the chitosan-tripolyphosphate nano-

    particle matrix was studied. Bovine serum albumin (BSA) was loaded into nanoparticles

    as a model drug. The final particle size, polydispersity index, entrapment efficiency and

    the release rate of BSA were optimized in relation to variations in parameters such as

    sodium alginate concentration, theoretical loading of BSA and degree of deacetylation

    of chitosan. It was found that increases in sodium alginate concentration resulted in

    smaller particle size, higher polydispersity index, lower entrapment efficiency and lower

    release rate of BSA. The same parametric changes were observed for the theoretical

    loading of BSA, with the exception of drug release which was followed by higher rate.

    Nanoparticles of chitosan-tripolyphosphate with a high degree of deacetylation led to

    production of smaller particle size, higher polydispersity index and entrapment

    efficiency, and a faster drug-release profile. The experimental data of this study

    revealed that burst release decreased significantly in reinforced chitosan-tripoly-

    phosphate nanoparticles.

    INTRODUCTION

    Application of chitosan, a cationicpolysaccharide, has been exten-

    sively investigated in various fields

    of applications, such as biotechno-

    logy [1], as a pharmaceutical

    excipient in oral drug formulations

    [2], waste water treatment [3], cos-

    metics [4] and food science [5].

    These broad fields of applications

    are due to chitosan's unique set of

    properties. Favourable biologicalproperties [6], low toxicity [7],

    biodegradability [8], proper

    mucoadhesive properties [9], abs-

    orption enhancer across intestinalepithelium and mucosal sites for

    drugs, peptides and proteins [10],

    metal complexation [11], antimi-

    crobial activity [12], good homo-

    static properties [13], and accept-

    able anti-cancerous properties [14]

    are the most important set of prop-

    erties. Among all applications of

    chitosan drug delivery systems are

    widely recognized as a promisingresearch field [15]. Tablets, micro-

    spheres, microgels and nano-

    particles are the different forms

    Available online at: http://journal.ippi.ac.ir

    http://www.sid.ir/
  • 7/29/2019 Iranian Journal Chitosan

    2/12

    of chitosan-based drug delivery devices that have

    been studied. In the past decade, chitosan nano-

    particles have been extensively investigated because

    they can control drug release rate, prolong the

    duration of therapeutic effectiveness, and deliverdrugs to their specific sites in the body [16]. Chitosan

    nanoparticles exhibit superior activity that can be

    attributed to their small and quantum size

    effect [17].

    Common methods to prepare chitosan nano-

    particles are ionic gelation, coacervation or precipi-

    tation, emulsion-droplet coalescence, reverse

    micellar, and self-assembly chemical modification

    [14]. The ionic gelation process is commonly used to

    prepare chitosan nanoparticles because it is a verysimple and mild method [18]. This process can be

    performed either by chemical or physical cross-

    linking. It is worth noting that chitosan has a high

    density of amine groups in its backbone and the

    amine groups are protonized to form -NH3+ in acidic

    solution. These positively charged groups in chitosan

    can be chemically cross-linked with dialdehydes such

    as glutaraldehyde [19] and ethylene glycol diglycidyl

    ether [20], or physically cross-linked with multivalent

    anions derived from sodium tripolyphosphate (TPP)

    [21], citrate [22,23] and sulphate [24]. Both

    glutaraldehyde and ethylene glycol diglycidyl ether

    are toxic and can cause irritation to mucosal

    membranes [25]. Physically cross-linked chitosan

    gels have been used in drug delivery systems due to

    their enhanced biocompatibility over chemically

    cross-linked chitosan [26].

    Non-toxicity and quick gelling ability of TPP are

    the important properties that make it a favourable

    cross-linker for ionic gelation of chitosan [14]. In

    addition, TPP has been also recognized as an

    acceptable food additive by the US Food and Drug

    Administration [27]. Moreover, the process of ionic

    gelation of chitosan with TPP as a cross-linker is

    feasible for the scale-up of entrapment in a particle

    processing operation [28]. Chitosan nanoparticles

    prepared by TPP as an anionic cross-linker are

    homogeneous, and possess positive surface charge

    that make them suitable for mucosal adhesion

    applications [14].

    The properties of ionically cross-linked chitosanare influenced by electrostatic interactions between

    the anionic cross-linker and chitosan. This interaction

    depends on the variables such as: anionic molecular

    structure, its charge density and molecular concen-

    tration, pH of chitosan solution, and physical

    properties of chitosan, i.e., molecular weight anddegree of deacetylation (DDA) [25]. Several studies

    have investigated the effect of these variables on the

    properties of ionically produced chitosan, its drug

    entrapment efficiency and drug release behaviour

    [25].

    Advantages of protein delivery systems based on

    chitosan-TPP nanoparticles are reported in many

    research works performed in the past few years [29].

    A shortcoming of chitosan-based nanoparticles as

    protein release system is that such particles release30-70% of the protein within 3-6 h placed in release

    environment. This is due to the mechanism of burst

    release that has been considered as a slow release in

    many studies [30].

    Burst release management is a major challenge in

    the development of drug delivery systems because it

    may lead to inefficient delivery and significant

    toxicity hazards. The degree of burst release general-

    ly depends upon the nature of the polymer, drug

    molecular structure and its molecular weight, poly-

    mer/drug ratio, relative affinities of the drug and

    polymer and the aqueous phase [31].

    Many methods, including blending with other

    polymers, grafting with monomers, varying the

    chemistry of the polymer [32], adding excipients into

    polymer phase [33], employing new polymers [34],

    encapsulating particulate forms of the drugs into

    microparticles, preparation of novel biopolymer/

    inorganic material composites [35] and spray/

    freezing [36] have been applied to improve the burst

    release properties of the polymeric carriers.

    Polymer coating is another burst release control

    method. Tavakol et al. [37] have studied this method

    for N,O-carboxymethyl chitosan (NOCC) beads

    coated with chitosan. They produced beads of NOCC

    and alginate with ionotropic gelation method and

    then, coated the beads with chitosan. The effect of

    coating and drying methods on the swelling and

    release behaviour of the prepared beads has been

    investigated. The results of this work indicate that

    burst release has not been observed in chitosancoated beads. Using additional barrier layers is

    Reinforcement of Chitosan Nanoparticles Obtained ... Hasanzadeh Kafshgari M et al.

    Iranian Polymer Journal / Volume 20 Number 5 (2011)446

    http://www.sid.ir/
  • 7/29/2019 Iranian Journal Chitosan

    3/12

    naturally an efficient method to manipulate drug

    release profiles and reduce the burst release, though it

    is inherently an expensive method due to the

    additional materials employed and difficulties

    associated with controlling the barriers quality. Chenet al. [30] have reported nanoparticles based on

    N-trimethyl chitosan chloride (TMC) and TPP. They

    used BSA as a model protein entrapped into the

    particles with sodium alginate as a modifier of TMC

    nanoparticles. They showed that burst release of BSA

    can be reduced from 60% to 45%.

    The main objective of this work was to reduce the

    burst release of BSA-loaded chitosan nanoparticles

    prepared by a one-step simple method. Very few

    studies have focused on high loadings (10-15%) ofproteins in micro/nanospheres with low burst release.

    Due to the nature of BSA, an optimum BSA delivery

    system must undergo burst release as low as possible

    while keeping drug loading as high as possible. The

    design of an optimum system is the main objective in

    this study. Comparison of the release test results of the

    current work with other similar works clearly shows

    higher efficiency of BSA-loaded chitosan nano-

    particles by the method developed in this study. While

    this work and the study carried out by Chen et al. [30]

    are both using ionic gelation method, two important

    differences are to be taken into account. Chen et al.

    [30] usedN-trimethyl chitosan as a polymeric carrier

    without any ionotropic cross-linker. In this work

    chitosan is used as a polymeric carrier and CaCl2 as

    an ionotropic cross-linker to further reinforce the

    chitosan/alginate composite matrix. In order to

    decrease burst release, chitosan nanoparticles with

    two different degrees of deacetylation were modified

    using calcium alginate, and the effects of sodium

    alginate concentration on particle size, entrapment

    efficiency and BSA release profile were investigated.

    EXPERIMENTAL

    Materials

    Medium molecular weight chitosan (Fluka, The

    Netherlands) was used as an encapsulating polymer.

    According to its specification analysis, the molecular

    weight was 400 kDa. The degree of deacetylation wasabout 83% as determined by potentiometric titration.

    Alginic acid as its sodium salt (medium viscosity,

    3500 cps, 2% (w/v) aqueous solution at 25C from

    Sigma-Aldrich, The Netherlands) was used as a rein-

    forcing polymeric agent. Sodium tripolyphosphate as

    ionic cross-linker was supplied by Sigma-Aldrich.Calcium chloride was provided by Merck Chemical

    Co., Germany, and used as ionotropic cross-linking

    agent. As a model protein molecule for entrapment

    and control release studies, BSA was obtained from

    Merck Chemical Co., Germany. The other chemicals

    were analytical grade reagents and used as received.

    Purification of Chitosan

    The chitosan was dissolved in 2% (v/v) acetic acid

    solution which was filtered to remove the residues ofinsoluble particles. A two-molar solution of NaOH

    was added to the filtrate to obtain purified chitosan in

    the form white precipitate. The precipitated chitosan

    was washed thoroughly using double distilled water,

    and then dried at 40C for 48 h. Further deacetylation

    of chitosan occurred during the purification process.

    Determination of Chitosan Degree of Deacetylation

    The degree of deacetylation numerical values of the

    unpurified and purified chitosan samples were

    determined by potentiometric titration of chitosan

    (25 mL of 0.2 M HCl) against 0.1 M NaOH using the

    following equation [38]:

    (1)

    where, V is the volume of alkali consumed in the

    titration of amino groups present in the chitosan

    samples, CNaOH is the concentration of sodium

    hydroxide used for titration, MChitosan is the mass of

    chitosan sample, and 16 and 0.0994 are the molecular

    weight and theoretical molar mass of the amino

    groups, respectively.

    Preparation of Chitosan Nanoparticles

    Chitosan nanoparticles were prepared on the basis of

    ionic gelation of chitosan in presence of TPP.

    Unpurified and purified medium molecular weight

    chitosan were dissolved in 1% (v/v) acetic acid

    solution at the final concentration of 0.5% (w/v). Two

    different concentrations of 20% and 50% (w/w) BSA,based on chitosan and alginate, were added each into

    447Iranian Polymer Journal / Volume 20 Number 5 (2011)

    Reinforcement of Chitosan Nanoparticles Obtained ...Hasanzadeh Kafshgari M et al.

    0994.0

    1610(%)

    3

    =

    Chitosan

    NaOH

    M

    CVDDA

    http://www.sid.ir/
  • 7/29/2019 Iranian Journal Chitosan

    4/12

    a separate chitosan solution prior to nanoparticles

    formation. The pH of chitosan-BSA solution was

    adjusted to 5.9 by 2 N NaOH solution. Then, 10 mL

    of TPP solution (0.5% w/v) prepared in double

    distilled water was added into the chitosan-BSAsolution (50 mL) through an insulin syringe needle at

    the speed of 60 mL/h under magnetic stirring at room

    temperature. The pH of chitosan-BSA solution

    reached 5.5 after adding TPP solution. The procedure

    described so far results in producing unreinforced

    chitosan nanoparticles.

    Reinforced chitosan nanoparticles were prepared

    by the same procedure, except that various amounts

    of sodium alginate (10% and 20% w/v) were

    dissolved in the TPP solution before adding thechitosan-BSA solution. Chitosan nanoparticles were

    formed upon adding the TPP solution to chitosan-

    BSA solution and mixing. After ten minutes of

    mixing these two solutions, 0.9 g calcium chloride

    was added to the suspension which was stirred

    for 20 min. Finally, the nanoparticles were

    isolated by centrifugation at 6000 rpm for 45 min.

    The supernatant was stored for further analysis.

    The precipitate was suspended in double distilled

    water, centrifuged again and dried at 37C. The

    dried chitosan nanoparticles were suspended

    in milli-Q distilled water for characterization tests

    where appropriate; or directly used for in vitro

    Table 1. Formulations of the prepared chitosan-TPP

    samples loaded with BSA.

    release experiments.

    Formulations of all chitosan-TPP samples that

    were used to investigate the effect of BSA loading,

    alginate concentration, and degree of deacetylation

    on nanoparticles and in vitro release profiles arepresented in Table 1.

    Characterization Tests of Nanoparticles

    The chemical interactions between chitosan,

    alginate, TPP and BSA were established through

    FTIR spectrometry. FTIR Spectra of chitosan, TPP

    and BSA prepared nanoparticles were recorded on a

    460 Plus Jasco Inc FTIR spectrophotometer

    (Maryland, USA) within KBr pellets. The instrument

    was operated with resolution of 4 cm-1

    andfrequency range of 400-4000 cm-1.

    The mean particle size and size distribution of

    nanoparticles were determined by dynamic light

    scattering (DLS) using a Malvern Zetasizer Nano-

    ZS-ZEN 1600 (Malvern Instruments Co., UK). The

    analysis was carried out at a scattering angle of 90 at

    a temperature of 25C using nanoparticles dispersed

    in de-ionized distilled water. Every sample measure-

    ment was repeated 10 times. Particle size distribution

    of the nanoparticles is reported as a polydispersity

    index (PDI). The morphology of dried nanoparticles

    was observed by transmission electron microscopy

    (TEM). For TEM analysis, about 3 mL ethanol was

    added to the prepared chitosan nanoparticles, and

    dispersed in Sonicator (Power Sonic 405, Hwashin

    Technology Co., Korea) for 5 min, and one droplet of

    the nanoparticles solution was dripped onto copper

    grids. The samples were dried by natural air flow

    and then analyzed using TEM without any further

    treatment or coating.

    To determine the entrapment efficiency (EE) of

    BSA into nanoparticles, the amount of free BSA in

    supernatant was analyzed with UV-Vis spectro-

    photometer at 595 nm using the Bradford protein

    assay. The supernatant of non-loaded chitosan (DDA:

    83.5% or 96.4%) nanoparticle suspension was used

    as a blank. The BSA entrapment efficiency (EE) was

    calculated using the following equation:

    (2)

    Iranian Polymer Journal / Volume 20 Number 5 (2011)448

    Reinforcement of Chitosan Nanoparticles Obtained ... Hasanzadeh Kafshgari M et al.

    inBSAfreeaddedBSAtotalEE = [((%)

    100]/) addedBSAtotaltsupernatan

    Sample Alginate concentration

    (w/v%)

    Theoretical

    loading (w/w%)

    DDA

    (%)

    III

    III

    IV

    V

    VI

    VII

    VIII

    IX

    X

    XI

    XII

    0.00.1

    0.2

    0.0

    0.1

    0.2

    0.0

    0.1

    0.2

    0.0

    0.1

    0.2

    2020

    20

    50

    50

    50

    20

    20

    20

    50

    50

    50

    83.5483.54

    83.54

    83.54

    83.54

    83.54

    96.38

    96.38

    96.38

    96.38

    96.38

    96.38

    http://www.sid.ir/
  • 7/29/2019 Iranian Journal Chitosan

    5/12

    In Vitro Drug Release

    The dried BSA-loaded nanoparticles were resuspend-

    ed in 25 mL of 0.1 M phosphate buffer solution (PBS,

    pH 7.4), then incubated at 37C while stirred at

    100 rpm. At specific time intervals, 0.5 mL of thesample was removed and replaced with fresh PBS.

    The withdrawn samples were centrifuged at

    6000 rpm for 5 min. The BSA released from the

    nanoparticles and present in the supernatant was

    measured by Bradford protein assay. The blank

    sample consisted of non-loaded chitosan nano-

    particles resuspended in PBS. Triplicate samples were

    analyzed for each measurment.

    RESULTS AND DISCUSSION

    Chitosan TPP nanoparticles are formed by ionic

    cross-linking (ionic gelation) of oppositely charged

    ions of chitosan and TPP. However, the preparation of

    reinforced chitosan-TPP nanoparticles has been

    attributed to the three following phenomena: (a) ionic

    gelation technique between positively charged

    chitosan and negatively charged TPP and BSA, (b)

    electrostatic interaction between the positively

    charged -NH3+ of chitosan and negatively charged

    -COO- of alginate, and (c) ionotropic gelation process

    between negatively charged alginate and calcium ion

    (Ca+2) [39]. In this study, calcium ion is used as a

    cross-linking agent to strengthen and stabilize the

    particles.

    The ratio between chitosan and TPP is critical and

    does control the size and PDI of the nanoparticles. It

    is shown that by increasing the chitosan/TPP ratio,

    smaller nanoparticles can be produced [16]. TPP is a

    polyfunctional cross-linking agent and can create five

    ionic cross-linking points with amino groups of

    chitosan. Thus, in this study the ratio of 5/1 (w/w) was

    selected as a suitable chitosan/TPP ratio [16].

    The ionic interaction between chitosan and TPP is

    dependent on the solution pH. This is due to the

    variation in ionization degree of chitosan and TPP.

    Other researchers [25] showed that a pH range 3-6

    was a suitable range for ionic gelation of chitosan

    with TPP. The pH of 5.9 is selected in the present

    work because BSA (PI = 4.8) is negatively charged atthis pH and electrostatic interaction between the

    negatively charged BSA and positively charged

    chitosan causes greater entrapment of BSA into the

    nanoparticles.

    FTIR was used to confirm the incorporation of

    calcium alginate into the chitosan matrix and loadingof BSA in the prepared nanoparticles. The FTIR

    spectra of chitosan, alginate, BSA, BSA-loaded

    chitosan nanoparticle (sample IV) and BSA-loaded

    chitosan-alginate nanoparticles (sample VI) are

    presented in Figure 1.

    Three characteristics absorption bands observed in

    chitosan (spectrum a in Figure 1), at 3413, 1672 and

    1317 cm-1, as in the given order are due to the N-H,

    amide I and amide III groups present in chitosan.

    According to Yuan et al. [40] the peak of 3413 cm-1

    corresponds to stretching vibration of N-H in pure

    chitosan. It is noticeable that this peak has shifted to

    lower wavenumbers centered at 3395 cm-1 and it is

    broader and stronger in chitosan particles (samples IV

    and VI, spectra d and e in Figure 1). This is an

    evidence of molecular interaction between these

    Figure 1. FTIR Spectra of: (a) chitosan, (b) alginate, (c)

    BSA, (d) BSA-loaded chitosan nanoparticle (sample IV) and(e) BSA-loaded chitosan-alginate nanoparticle (sample VI).

    Reinforcement of Chitosan Nanoparticles Obtained ...Hasanzadeh Kafshgari M et al.

    Iranian Polymer Journal / Volume 20 Number 5 (2011) 449

    http://www.sid.ir/
  • 7/29/2019 Iranian Journal Chitosan

    6/12

    groups and sodium tripolyphosphate and also due to

    hydrogen bonding involvement. Primary amines also

    show sharp peak between 3500 and 3400 cm-1 which

    could be attributed to the asymmetric and symmetric

    stretching of the N-H bonds [40]. The peak in purechitosan and chitosan particles appears broad in this

    region due to the contribution of O-H stretching peaks

    and hydrogen bonding [41].

    FTIR Spectra of BSA showed peaks around 1655,

    1534 and 3309 cm-1, reflecting the acetylamino I,

    acetylamino II and (NH2) groups, respectively [42].

    Acetylamino I at 1655 cm-1 and acetylamino II at

    1534 cm-1 in BSA (spectrum c in Figure 1) overlapped

    1672 cm-1 of amide I and 1597 cm-1 in chitosan, so

    more intensive peaks appeared for the BSA-loadedchitosan nanoparticles (sample IV, spectrum d in

    Figure 1).

    The characteristic peaks of sodium alginate

    included O-H at 3397 cm-1, COO- (asymmetric) at

    1617, COO- (symmetric) at 1429 and 1028 cm-1 for

    C-O-C stretching. For the BSA-loaded chitosan

    nanoparticle of sample IV, the stretching vibration of

    -OH and -NH2 at 3392 cm-1 became broader.

    Spectrum e in Figure 1 is an indication of intra-

    molecular and intermolecular hydrogen bonds which

    were formed and enhanced between chitosan and

    alginate molecules [43].

    The mean particle size, PDI and EE of the

    prepared samples are presented in Table 2. PDI is a

    Table 2. Mean particle size and entrapment efficiency of

    chitosan-TPP nanoparticles loaded with BSA.

    measure of homogeneity in dispersed systems and

    ranges from 0 to 1. Homogeneous dispersion has PDI

    value close to zero while PDI values greater than 0.3

    suggest high heterogeneity [44]. Except the three

    samples (samples I, IV and XII) other prepared

    nanoparticles have PDI values lower than 0.3

    (Table 2).

    Morphological studies of nanoparticles were

    conducted by TEM. The TEM images such as those

    presented in Figure 2 indicate that nanoparticles are

    Reinforcement of Chitosan Nanoparticles Obtained ... Hasanzadeh Kafshgari M et al.

    450 Iranian Polymer Journal / Volume 20 Number 5 (2011)

    Sample Mean particle

    size (nm)

    PDI Entrapment

    efficiency (%)

    I

    II

    III

    IV

    V

    VI

    VII

    VIII

    IX

    X

    XI

    XII

    396

    116

    459

    615

    255

    459

    295

    255

    190

    459

    220

    122

    0.162

    0.572

    0.188

    0.179

    0.201

    0.226

    0.075

    0.123

    0.631

    0.274

    0.261

    0.523

    70.54

    71.55

    75.14

    60.97

    59.48

    55.4

    84.55

    75.21

    71.12

    80.74

    69.16

    68.56

    (a) (b)

    Figure 2. TEM Photographs of samples: (a) IV and (b) VI.

    http://www.sid.ir/
  • 7/29/2019 Iranian Journal Chitosan

    7/12

    not spherical in shape. The size of nanoparticles

    based on TEM was smaller than the size determined

    by DLS. This can be attributed to the dehydration of

    the nanoparticles in the process of sample preparation

    required for TEM. It is worth noting that DLS givesthe size of swollen particles. Swelling ratio of some

    selected samples of the nanoparticles was measured

    according to the method proposed by Denkbas et al.

    [45].

    About 0.050 g of the nanoparticles was inserted

    into a tube with a diameter of 5 mm with a height of

    100 mm. The level of the beads in the tube was

    marked before filling it with 1 mL of distilled water

    and the tube was left for 24 h. After 24 h, the height

    of the beads in the solution was measured. Thepercentage of swelling was calculated based on the

    following equation:

    (3)

    where S is the percentage of swelling, ht is the height

    of swollen beads (cm) at specific time t and ho is the

    initial height of the beads (cm).

    The obtained results for samples IV, V and VI

    based on three independent measurements for eachsample were 119.3% 20.3, 223% 67 and 146.3%

    18.8, respectively.

    Effect of Alginate Concentration on

    Physicochemical Properties of Nanoparticles

    As can be seen in Table 2, mean particle size

    decreases as sodium alginate concentration increases.

    By contrast, PDI of the prepared nanoparticles

    increases as more sodium alginate was added to the

    gelation system. In ionic gelation process, TPP as a

    cross-linking agent forms hydrogen bond with free

    amine groups of BSA and chitosan. Sodium alginate

    competes with TPP to form electrostatic complex

    with chitosan and BSA. While smaller mean particle

    size can be attributed to strong inter-chain reactions

    between chitosan and alginate, as more sodium

    alginate causes local aggregation and increases PDI

    [30]. In other words, the reduction in particle size

    determined by DLS at swollen state in presence of

    calcium alginate in the network is due to higher

    degree of cross-linking of the whole network. Higher

    concentration of sodium alginate results in reduced

    entrapment efficiency (Table 2). This may be due to

    competition of anionic alginate and BSA to form

    interchain bonds with cationic chitosan.

    BSA Loading in Relation to the Physicochemical

    Properties of Nanoparticles

    The general pattern which can be deduced from the

    experimental data given in Table 2 is that, lowering

    theoretical loading of BSA leads to formation of

    smaller nanoparticles. BSA loading of nanoparticles

    is based on two different phenomena such as

    entrapment into particles and adsorption on the

    particle surface. As mentioned earlier, BSA

    molecules are negatively charged under the solution

    conditions used in this study. BSA entrapment intoparticles is due to interactions between oppositely

    charged chitosan and BSA, and formation of

    hydrogen bonds between the TPP and BSA.

    Additional adsorption of BSA on particle surface may

    also occur [14] that leads to higher mean particle size.

    PDI of the prepared nanoparticles increases with

    theoretical BSA of higher loading. Also, the increase

    of the particle size and PDI by increased BSA can be

    attributed to the increased viscosity of the chitosan/

    BSA solution and ionic interaction, respectively. Fewdata from Table 2 regarding the effect of BSA loading

    on entrapment efficiency of nanoparticles are

    presented in Figure 3. Sample preparation for

    samples I and IV were the same except for the

    theoretical BSA loading that was higher for sample

    IV compared to sample I. The same sample prepara-

    tion was applied to sample pair VIII and XI. It can be

    Figure 3. Effect of theoretical BSA loading on entrapment

    efficiency.

    Reinforcement of Chitosan Nanoparticles Obtained ...Hasanzadeh Kafshgari M et al.

    Iranian Polymer Journal / Volume 20 Number 5 (2011) 451

    100

    =

    o

    ot

    h

    hhS

    http://www.sid.ir/
  • 7/29/2019 Iranian Journal Chitosan

    8/12

    concluded from Figure 3 that higher theoretical BSA

    loading results in lowering entrapment efficiency. It is

    worth noting that data from Table 2 other than those

    presented in Figure 3 also support this conclusion.

    The decrease of entrapment efficiency with initiallyincreased drug content is due to increased chemical

    potential of the drug in chitosan/BSA solution. This

    conclusion is in agreement with reported studies such

    as the one carried out by Xu et al. [46] and it is in con-

    trast to other research reports attesting the opposite

    effect of drug loading on entrapment efficiency [14].

    DDA in Relation to the Physicochemical Properties

    of Nanoparticles

    The effects of degree of deacetylation on particle sizeand entrapment efficiency of BSA were investigated

    and the obtained experimental data are presented in

    Table 2. The data show that samples which were

    prepared with higher DDA chitosan (purified

    chitosan, DDA 96.4%) have smaller mean particle

    size than those prepared with unpurified chitosan

    (DDA 83.5%).

    In this study, purification process was conducted

    by NaOH. It is anticipated that further deacetylation

    of chitosan molecules may have occurred under

    strong caustic condition. This means that more amine

    groups are presented in purified chitosan of high

    DDA, which can be transformed into -NH3+ at acidic

    solution. The ionic gelation process is dependent on

    the number of positively charged groups (-NH3+) left

    on the dissolved chitosan molecules. Due to sufficient

    positively charged groups, the process of nano-

    particles production occurs more easily for chitosan

    with high DDA leading to smaller particles [47].

    Entrapment of negatively charged BSA molecules is

    also occurring more efficiently in the presence of

    positively charged amine groups. Sample preparation

    for samples I and VII was the same except for the

    DDA that was higher for sample VII compared to

    sample I. The same sample preparation is applicable

    to sample pairs II-VIII, III-IX, IV-X, V-XI and VI-

    XII. The presented data in Table 2 for these pairs

    reveal that entrapment efficiency of BSA increases at

    higher DDA.

    In Vitro Release StudyExperimental data on BSA release studies are

    presented in Figure 4. The data show that both BSA

    release rate and especially burst release are reduced

    significantly with higher sodium alginate concentra-

    tion used in preparation of nanoparticles. Sustained

    drug release from the nanoparticles is important inmany fields of applications. BSA release follows a

    biphasic pattern, characterized by initial burst release

    followed by a period of slower release. According to

    the presented data in Figure 4, fractional BSA

    released for sample IV (chitosan-TPP nanoparticles

    without alginate) and sample VI (reinforced chitosan-

    TPP with alginate) in 30 min are 10.5% and 3.5%,

    respectively. Also, these quantities in 6 h reach 14%

    and 11.8% for samples IV and VI, respectively. In

    addition, fractional drug released for sample IV is35.7% at 98 h while this quantity for sample VI is

    26.8% at 98 h. These results prove that reinforcement

    of chitosan-TPP nanoparticles with CaCl2 can

    reduce both burst release and release rate of BSA.

    Reinforcement of chitosan-TPP matrix with

    calcium alginate may be considered the cause for

    lower burst effect and slower release rate.

    Modification of N-trimethyl chitosan chloride

    (TMC)-TPP matrix by sodium alginate studied by

    Chen et al. [30] also showed that sodium alginate

    demonstrates the same result on burst effect and

    release rate as calcium alginate in this study. The

    amine groups in chitosan interact with TPP through

    ionic bonding. The interaction between chitosan

    matrix and alginate and ionotropic gelation of sodium

    alginate with CaCl2 in reinforced nanoparticles may

    Figure 4. Effect of sodium alginate concentration on BSA

    release profile from samples: () IV, () VI and (S) XIII.Release medium: T = 37C0.2, pH 7.4.

    452 Iranian Polymer Journal / Volume 20 Number 5 (2011)

    Reinforcement of Chitosan Nanoparticles Obtained ... Hasanzadeh Kafshgari M et al.

    http://www.sid.ir/
  • 7/29/2019 Iranian Journal Chitosan

    9/12

    also contribute to enhanced cross-linking density of

    the matrix. Increased cross-linking density may lead

    to lower diffusion of BSA from the reinforced matrix

    which consequently decreases the burst release and

    rate of release. To investigate the effect of sodiumalginate on drug release profile, without its

    conversion to calcium alginate network, a new sample

    designated as XIII, was prepared. Preparation of

    samples XIII and VI were the same except that CaCl2was not used in preparation of sample XIII. The

    release profile of this sample is shown in Figure 4. As

    can be seen, the burst release and release rate of BSA

    is lower in sample VI compared to sample XIII.

    Fractional BSA release profiles for two theoretical

    BSA loadings, 20% and 50% corresponding toloading capacity of 4.13% 0.04 and 9.75% 0.32,

    are presented in Figure 5. Release profiles show that

    when loading capacity of BSA increases from 4.13%

    0.04 to 9.75% 0.32, total BSA release at 6 h

    increases from 7.5% to 15.8%. The nanoparticles

    prepared with higher loading capacity of BSA have

    greater overall release. At higher BSA loading, BSA

    binding on chitosan molecule is weaker, and some of

    the loaded BSA molecules are adsorbed onto the

    surface of particles. Weak binding and adsorption of

    BSA on the particle surfaces leads to higher release

    rate. This result is in agreement with reports by other

    researchers [14].

    Effect of DDA on the release profile is shown in

    Figure 6. The results show that BSA release rate and

    burst release increase with higher DDA. As it was

    Figure 5. Effect of theoretical BSA loading on BSA release

    profile from samples: () IX and () XII. Release medium:T = 37C0.2, pH 7.4.

    Figure 6. Effect of chitosan degree of deacetylation on BSA

    release profile from samples: () VI and () XII. Release

    medium: T = 37C0.2, pH 7.4.

    stated above, higher DDA causes significant

    reduction in mean particle size, thus increasing the

    ratio of surface area to volume of nanoparticles, that

    consequently increases the release rate and burst

    release.

    CONCLUSION

    Reinforced chitosan-TPP nanoparticles loaded with

    BSA were prepared by ionic gelation. Calcium

    alginate was used as a reinforcing agent. Size of the

    prepared nanoparticles ranged from 116 to 615 nm

    with PDI lower than 0.3. TEM Analysis demonstrated

    that nanoparticles were not spherical. At higher

    sodium alginate concentration smaller mean particle

    size was obtained. High concentration of sodium

    alginate solution resulted in lower entrapment

    efficiency. Higher degree of deacetylated chitosan

    lowered particle size and improved the entrapment

    efficiency. This study showed that reinforcement of

    chitosan-TPP nanoparticles with calcium alginate sig-

    nificantly lowered the burst release and consequently

    the BSA release rate. Increased theoretical loading of

    BSA and degree of deacetylation of chitosan led to

    increased burst release and release rate.

    ACKNOWLEDGEMENT

    The authors are grateful to the financial supports from

    Reinforcement of Chitosan Nanoparticles Obtained ...Hasanzadeh Kafshgari M et al.

    Iranian Polymer Journal / Volume 20 Number 5 (2011) 453

    http://www.sid.ir/
  • 7/29/2019 Iranian Journal Chitosan

    10/12

    the Vice-Chancellery for Research and Technology

    Affairs of University of Sistan and Baluchestan.

    REFERENCES

    1. Liu X, Yang F, Song T, Zeng A, Wang Q, Sun Z,

    Shen J, Effects of chitosan, O-carboxymethyl

    chitosan and N-[(2-hydroxy-3-N,N-dimethylhexa-

    decyl ammonium) propyl] chitosan chloride on

    lipid metabolism enzymes and low-density-

    lipoprotein receptor in a murine diet-induced

    obesity, Carbohyd Polym, 85, 334-340, 2011.

    2. Baldrick P, The safety of chitosan as a pharmaceu-

    tical excipient,Regul Toxicol Pharmacol, 56, 290-299, 2010.

    3. Gerente C, Andres Y, McKay G, Le Cloirec P,

    Removal of arsenic(V) onto chitosan: from

    sorption mechanism explanation to dynamic water

    treatment process, Chem Eng J, 158, 593-598,

    2010.

    4. Harris R, Lecumberri E, Mateos-Aparicio I,

    Mengibar M, Heras A, Release characteristics of

    vitamin E incorporated chitosan microspheres and

    in vitro-in vivo evaluation for topical application,

    Carbohyd Polym, 84, 803-806, 2011.

    5. Fernandez-Saiz P, Ocio MJ, Lagaron JM,

    Antibacterial chitosan-based blends with ethylene-

    vinyl alcohol copolymer, Carbohyd Polym, 80,

    874-884, 2010.

    6. Alves NM, Mano JF, Chitosan derivatives

    obtained by chemical modifications for biomedical

    and environmental applications, Int J Biolog

    Macromol, 43, 401-414, 2008.

    7. Gao J-Q, Zhao Q-Q, Lv T-F, Shuai W-P, Zhou J,

    Tang G-P, Liang W-Q, Tabata Y, Hu Y-L, Gene-

    carried chitosan-linked-PEI induced high gene

    transfection efficiency with low toxicity and

    significant tumor-suppressive activity, Int J

    Pharm, 387, 286-294, 2010.

    8. Bagheri-Khoulenjani S, Taghizadeh SM, Mirzadeh

    H, An investigation on the short-term biodegrad-

    ability of chitosan with various molecular weights

    and degrees of deacetylation,Carbohyd Polym, 87,

    773-778, 2009.

    9. Patil SB, Sawant KK, Chitosan microspheres as adelivery system for nasal insufflation, Colloid Surf

    B: Biointerfaces, 84, 384-389, 2011.

    10. Van der Lubben IM, Verhoef JC, Borchard G,

    Junginger HE, Chitosan for mucosal vaccination,

    Adv Drug Delivery Rev, 52, 139-144, 2001.

    11. Trimukhe KD, Varma AJ, A morphological studyof heavy metal complexes of chitosan and

    crosslinked chitosans by SEM and WAXRD,

    Carbohyd Polym, 71, 698-702, 2008.

    12. Kong M, Chen XG, Xing K, Park HJ,

    Antimicrobial properties of chitosan and mode of

    action: a state of the art review, Int J Food

    Microbiol, 144, 51-63, 2010.

    13. Ong S-Y, Wu J, Moochhala SM, Tan M-H, Lu J,

    Development of a chitosan-based wound dressing

    with improved hemostatic and antimicrobialproperties,Biomaterials, 29, 4323-4332, 2008.

    14. Gan Q, Wang, T, Chitosan nanoparticle as protein

    delivery carrier-systematic examination of fabri-

    cation conditions for efficient loading and release,

    Colloid Surf B: Biointerfaces, 59, 24-34, 2007.

    15. Rao KSVK, Rao KM, Kumar PVN, Chung I-D,

    Novel chitosan-based pH sensitive micro-net-

    works for the controlled release of 5-fluorouracil,

    Iran Polym J, 19, 265-276, 2010.

    16. Papadimitriou S, Bikiaris D, Avgoustakis K,

    Karavas E, Georgarakis M, Chitosan nanoparti-

    cles loaded with dorzolamide and pramipexole,

    Carbohyd Polym, 73, 44-54, 2008.

    17. Qi L, Xu Z, Jiang X, Hu C, Zou X, Preparation

    and antibacterial activity of chitosan nano-

    particles, Carbohyd Res, 339, 2693-2700, 2004.

    18. Tsai ML, Bai SW, Chen RH, Cavitation effects

    versus stretch effects resulted in different size and

    polydispersity of ionotropic gelation chitosan-

    sodium tripolyphosphate nanoparticle, Carbohyd

    Polym, 71, 448-457, 2008.

    19. Xiong W-W, Wang W-F, Zhao L, Song Q, Yuan

    L-M, Chiral separation of (R,S)-2-phenyl-1-

    propanol through glutaraldehyde-crosslinked

    chitosan membranes,J Membr Sci, 328, 268-272,

    2009.

    20. Azlan K, Saime WNW, Laiken L, Chitosan and

    chemically modified chitosan beads for acid dyes

    sorption,J Environ Sci, 21, 296-302, 2009.

    21. Liu H, Gao C, Preparation and properties of

    ionically cross-linked chitosan nanoparticles,Polym Adv Technol, 20, 613-619, 2009.

    Reinforcement of Chitosan Nanoparticles Obtained ... Hasanzadeh Kafshgari M et al.

    454 Iranian Polymer Journal / Volume 20 Number 5 (2011)

    http://www.sid.ir/
  • 7/29/2019 Iranian Journal Chitosan

    11/12

    22. Chen S, Liu M, Jin S, Wang B, Preparation of

    ionic-crosslinked chitosan-based gel beads and

    effect of reaction conditions on drug release

    behaviors,Int J Pharm, 349, 180-187, 2008.

    23. Varshosaz J, Alinagari R, Effect of citric acid ascross-linking agent on insulin loaded chitosan

    microspheres,Iran Polym J, 14, 647-656, 2005.

    24. Zhong Z, Li P, Xing R, Liu S, Antimicrobial

    activity of hydroxylbenzenesulfonailides deriva-

    tives of chitosan, chitosan sulfates and car-

    boxymethyl chitosan,Int J Biolog Macromol, 45,

    163-168, 2009.

    25. Gupta KC, Jabrail FH, Controlled-release

    formulations for hydroxyl urea and rifampicin

    using polyphosphate-anion-crosslinked chitosanmicrospheres,J Appl Polym Sci, 104, 1942-1956,

    2007.

    26. Rayment P, Butler MF, Investigation of ionically

    crosslinked chitosan and chitosan-bovine serum

    albumin beads for novel gastrointestinal func-

    tionality, J Appl Polym Sci, 108, 2876-2885,

    2008.

    27. Lin Y-H, Sonaje K, Lin KM, Juang J-H, Mi F-L,

    Yang H-W, Sung H-W, Multi-ion-crosslinked

    nanoparticles with pH-responsive characteristics

    for oral delivery of protein drugs,J Control Rel,

    132, 141-149, 2008.

    28. Stulzer HK, Tagliari MP, Parize AL, Silva MAS,

    Laranjeira MCM, Evaluation of cross-linked

    chitosan microparticles containing acyclovir

    obtained by spray-drying,Mater Sci Eng: C, 29,

    387-392, 2009.

    29. Rekha MR, Sharma CP, Synthesis and evaluation

    of lauryl succinyl chitosan particles towards oral

    insulin delivery and absorption, J Control Rel,

    135, 144-151, 2009.

    30. Chen F, Zhang Z-R, Huang Y, Evaluation and

    modification ofN-trimethyl chitosan chloride

    nanoparticles as protein carriers, Int J Pharm,

    336, 166-173, 2007.

    31. Sheikh Hassan A, Sapin A, Lamprecht A, Emond

    E, El Ghazouani F, Maincent P, Composite

    microparticles with in vivo reduction of the burst

    release effect,Eur J Pharm Biopharm, 73, 337-

    344, 2009.

    32. Desai KGH, Mallery SR, Schwendeman SP,Effect of formulation parameters on 2-

    methoxyestradiol release from injectable

    cylindrical poly(dl-lactide-co-glycolide)

    implants, Eur J Pharm Biopharm, 70, 187-198,

    2008.

    33. Katare YK, Panda AK, Influences of excipientson in vitro release and in vivo performance of

    tetanus toxoid loaded polymer particles, Eur J

    Pharm Sci, 28, 179-188, 2006.

    34. Cornejo-Bravo JM, Flores-Guillen ME, Lugo-

    Medina E, Licea-Claverie A, Drug release from

    complexes with a series of poly(carboxyalkyl

    methacrylates), a new class of weak poly-

    electrolytes,Int J Pharm, 305, 52-60, 2005.

    35. Zhang J, Wang Q, Wang A, In situ generation of

    sodium alginate/hydroxyapatite nanocompositebeads as drug-controlled release matrices,Acta

    Biomaterialia, 6, 445-454, 2010.

    36. Leach WT, Simpson DT, Val TN, Anuta EC, Yu

    Z, Williams RO, Johnston KP, Uniform encapsu-

    lation of stable protein nanoparticles produced by

    spray freezing for the reduction of burst release,

    J Pharm Sci, 94, 56-69, 2005.

    37. Tavakol M, Vasheghani-Farahani E, Dolatabadi-

    Farahani T, Hashemi-Najafabadi S, Sulfasalazine

    release from alginate-N,O-carboxymethyl chi-

    tosan gel beads coated by chitosan, Carbohyd

    Polym, 77, 326-330, 2009.

    38. Jiang X, Chen L, Zhong W, A new linear poten-

    tiometric titration method for the determination

    of deacetylation degree of chitosan, Carbohyd

    Polym, 54, 457-463, 2003.

    39. Li XY, Kong XY, Shi S, Zheng XL, Guo G, Wei

    YQ, Qian ZY, Preparation of alginate coated

    chitosan microparticles for vaccine delivery,

    BMC Biotechnol, 8, 89, 2008.

    40. Yuan Q, Shah J, Hein S, Misra RDK, Controlled

    and extended drug release behavior of chitosan-

    based nanoparticle carrier,Acta Biomaterialia, 6,

    1140-1148, 2010.

    41. Zhang L, Kosaraju SL, Biopolymeric delivery

    system for controlled release of polyphenolic

    antioxidants,Eur Polym J, 43, 2956-2966, 2007.

    42. Wan A, Sun Y, Li H, Characterization of novel

    quaternary chitosan derivative nanoparticles

    loaded with protein,J Appl Polym Sci, 114, 2639-

    2647, 2009.43. Dai Y-N, Li P, Zhang J-P, Wang A-Q, Wei Q, A

    Reinforcement of Chitosan Nanoparticles Obtained ...Hasanzadeh Kafshgari M et al.

    Iranian Polymer Journal / Volume 20 Number 5 (2011) 455

    http://www.sid.ir/
  • 7/29/2019 Iranian Journal Chitosan

    12/12

    novel pH sensitive N-succinyl chitosan/alginate

    hydrogel bead for nifedipine delivery,Biopharm

    Drug Dispos, 29, 173-184, 2008.

    44. Zhang L, Kosaraju SL, Biopolymeric delivery

    system for controlled release of polyphenolicantioxidants,Eur Polym J, 43, 2956-2966, 2007.

    45. Denkbas EB, Odabas IM, Chitosan microspheres

    and sponges: preparation and characterization,J

    Appl Polym Sci, 76, 1637-1643, 2000.

    46. Xu Y, Du Y, Effect of molecular structure of

    chitosan on protein delivery properties of chi-

    tosan nanoparticles,Int J Pharm, 250, 215-226,

    2003.

    47. Wang X, Chi N, Tang X, Preparation of estradiol

    chitosan nanoparticles for improving nasalabsorption and brain targeting, Eur J Pharm

    Biopharm, 70, 735-740, 2008.

    Reinforcement of Chitosan Nanoparticles Obtained ... Hasanzadeh Kafshgari M et al.

    456 Iranian Polymer Journal / Volume 20 Number 5 (2011)

    http://www.sid.ir/