Sei sulla pagina 1di 24

Journal of Controlled Release 100 (2004) 5 – 28

www.elsevier.com/locate/jconrel

Review

Recent advances on chitosan-based micro- and nanoparticles


in drug deliveryB
Sunil A. Agnihotri, Nadagouda N. Mallikarjuna, Tejraj M. Aminabhavi*
Drug Delivery Division, Center of Excellence in Polymer Science, Karnatak University, Dharwad 580 003, India
Received 15 July 2004; accepted 12 August 2004

Abstract

Considerable research efforts have been directed towards developing safe and efficient chitosan-based particulate drug
delivery systems. The present review outlines the major new findings on the pharmaceutical applications of chitosan-based
micro/nanoparticulate drug delivery systems published over the past decade. Methods of their preparation, drug loading, release
characteristics, and applications are covered. Chemically modified chitosan or its derivatives used in drug delivery research are
discussed critically to evaluate the usefulness of these systems in delivering the bioactive molecules. From a literature survey, it
is realized that research activities on chitosan micro/nanoparticulate systems containing various drugs for different therapeutic
applications have increased at the rapid rate. Hence, the present review is timely.
D 2004 Elsevier B.V. All rights reserved.

Keywords: Microparticles; Nanoparticles; Chitosan; Chemically modified chitosan; Drug delivery

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
2. Methods of preparation of micro/nanoparticles of chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
2.1. Emulsion cross-linking . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
2.2. Coacervation/precipitation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
2.3. Spray-drying . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
2.4. Emulsion-droplet coalescence method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
2.5. Ionic gelation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
2.6. Reverse micellar method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
2.7. Sieving method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15

B
This paper is CEPS Communication # 23.
* Corresponding author. Tel.: +91 836 2779983; fax: +91 836 2771275.
E-mail address: aminabhavi@yahoo.com (T.M. Aminabhavi).

0168-3659/$ - see front matter D 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.jconrel.2004.08.010
6 S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28

3. Drug loading into micro/nanoparticles of chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15


4. Drug release and release kinetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
5. Pharmaceutical applications of chitosan particulate systems . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
5.1. Colon targeted drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
5.2. Mucosal delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
5.3. Cancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
5.4. Gene delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
5.5. Topical delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
5.6. Ocular delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
5.7. Chitosan as a coating material . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
6. Chemically modified chitosans . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24

1. Introduction free amino and N-acetyl groups [9]. Usually 1–3%


aqueous acetic acid solutions are used to solubilize CS.
Chitosan (CS) is a polysaccharide, similar in Chitosan is biocompatible with living tissues since it
structure to cellulose. Both are made by linear h- does not cause allergic reactions and rejection. It breaks
(1Y4)-linked monosaccharides [see Fig. 1 (a)]. down slowly to harmless products (amino sugars),
However, an important difference to cellulose is that which are completely absorbed by the human body
CS is composed of 2-amino-2-deoxy-h-d-glucan [10]. Chitosan degrades under the action of ferments, it
combined with glycosidic linkages. The primary is nontoxic and easily removable from the organism
amine groups render special properties that make CS without causing concurrent side reactions. It possesses
very useful in pharmaceutical applications. Compared antimicrobial property and absorbs toxic metals like
to many other natural polymers, chitosan has a mercury, cadmium, lead, etc. In addition, it has good
positive charge and is mucoadhesive [1]. Therefore, adhesion, coagulation ability, and immunostimulating
it is used extensively in drug delivery applications [2– activity.
6]. Chitosan is obtained from the deacetylation of If degree of deacetylation and molecular weight of
chitin, a naturally occurring and abundantly available CS can be controlled, then it would be a material of
(in marine crustaceans) biocompatible polysaccharide. choice for developing micro/nanoparticles. Chitosan
However, applications of chitin are limited compared has many advantages, particularly for developing
to CS because chitin is structurally similar to cellulose, micro/nanoparticles. These include: its ability to
but chemically inert. Acetamide group of chitin can be control the release of active agents, it avoids the use
converted into amino group to give CS, which is of hazardous organic solvents while fabricating
carried out by treating chitin with concentrated alkali particles since it is soluble in aqueous acidic solution,
solution. Chitin and CS represent long-chain polymers it is a linear polyamine containing a number of free
having molecular mass up to several million Daltons. amine groups that are readily available for cross-
Chitosan is relatively reactive and can be produced in linking, its cationic nature allows for ionic cross-
various forms such as powder, paste, film, fiber, etc. linking with multivalent anions, it has mucoadhesive
[7,8]. Commercially available CS has an average character, which increases residual time at the site of
molecular weight ranging between 3800 and 20,000 absorption, and so on. Chitin and CS have very low
Daltons and is 66% to 95% deacetylated. toxicity; LD50 of CS in laboratory mice is 16 g/kg
Chitosan, being a cationic polysaccharide in neutral body weight, which is close to sugar or salt. Chitosan
or basic pH conditions, contains free amino groups and is proven to be safe in rats up 10% in the diet [11].
hence, is insoluble in water. In acidic pH, amino groups Various sterilization methods such as ionizing radia-
can undergo protonation thus, making it soluble in tion, heat, steam and chemical methods can be
water. Solubility of CS depends upon the distribution of suitably adopted for sterilization of CS in clinical
S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28 7

been used in the preparation of mucoadhesive


formulations [19–22], improving the dissolution rate
of the poorly soluble drugs [14,23,24], drug targeting
[25,26] and enhancement of peptide absorption
[20,21,27].
Many reports are available on the preparation of CS
microspheres [23,25,26,28,29]. Many methods used in
the development of microparticulate polymeric drug
delivery devices can also be used to prepare CS
microspheres [30–35]. Dodane and Vilivalam [3]
reviewed new approaches on pharmaceutical applica-
tions of CS and discussed its mechanisms of action in
various in vitro and in vivo models. Recent reviews
[36,37] addressed the issues on biomedical, pharma-
ceutical and biological aspects of chitin, CS and their
derivatives. Chitosan and its derivatives as a non-viral
vector for gene delivery [38] and CS-based gastro-
intestinal delivery systems [39] have been discussed.
Fig. 1. (a) Structure of chitosan [poly (h1– 4-d-glucosamine)]. (b)
Structure of cross-linked chitosan.
The recent review by Sinha et al. [40] covers various
methods of preparation and evaluation of CS micro-
applications [12]. In view of the above-mentioned spheres, but no attempt has been made to discuss
properties, CS is extensively used in developing drug nanoparticulate CS systems. Different types of CS-
delivery systems [7,8,13–18]. Particularly, CS has based drug delivery systems are summarized in Table 1.

Table 1
Chitosan-based drug delivery systems prepared by different methods for various kinds of drugs
Type of system Method of preparation Drug
Tablets matrix diclofenac sodium, pentoxyphylline, salicylic acid, theophylline
coating propranolol HCl
Capsules capsule shell insulin, 5-amino salicylic acid
Microspheres/Microparticles emulsion cross-linking theophylline, cisplatin, pentazocine, phenobarbitone, theophylline,
insulin, 5-fluorouracil, diclofenac sodium, griseofulvin, aspirin,
diphtheria toxoid, pamidronate, suberoylbisphosphonate,
mitoxantrone, progesterone
coacervation/precipitation prednisolone, interleukin-2, propranolol-HCl
spray-drying cimetidine, famotidine, nizatidine, vitamin D-2, diclofenac
sodium, ketoprofen, metoclopramide-HCl, bovine serum albumin,
ampicillin, cetylpyridinium chloride, oxytetracycline, betamethasone
ionic gelation felodipine
sieving method clozapine
Nanoparticles emulsion-droplet coalescence gadopentetic acid
coacervation/precipitation DNA, doxorubicin
ionic gelation insulin, ricin, bovine serum albumin, cyclosporin A
reverse micellar method doxorubicin
Beads coacervation/precipitation adriamycin, nifedipine, bovine serum albumin, salbutamol
sulfate, lidocaine– HCl, riboflavin
Films solution casting isosorbide dinitrate, chlorhexidine gluconate, trypsin,
granulocyte-macrophage colony-stimulating factor, acyclovir,
riboflavine, testosterone, progesterone, beta-oestradiol
Gel cross-linking chlorpheniramine maleate, aspirin, theophylline, caffeine,
lidocaine– HCl, hydrocortisone acetate, 5-fluorouracil
8 S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28

However, the micro/nanoparticulate drug delivery


systems offer numerous advantages over the conven-
tional dosage forms. These include improved efficacy,
reduced toxicity and improved patient compliance
[35,41–43]. The present review addresses the recent
trends in the area of micro/nanoparticulate CS-based
drug delivery systems. Literature of the past decade has
been covered and results are critically evaluated.

2. Methods of preparation of micro/nanoparticles


of chitosan

Different methods have been used to prepare CS


particulate systems. Selection of any of the methods
depends upon factors such as particle size require-
Fig. 2. Schematic representation of preparation of chitosan
ment, thermal and chemical stability of the active
particulate systems by emulsion cross-linking method.
agent, reproducibility of the release kinetic profiles,
stability of the final product and residual toxicity since it involves tedious procedures as well as use of
associated with the final product. Different methods harsh cross-linking agents, which might possibly
used in the preparation of CS micro/nanoparticles are induce chemical reactions with the active agent.
discussed in this review. However, selection of any of However, complete removal of the un-reacted cross-
these methods depends upon the nature of the active linking agent may be difficult in this process.
molecule as well as the type of the delivery device. Recently, [33] we have used the emulsion cross-
Since we are concerned only with the micro/nano- linking method to prepare chitosan microspheres to
particulate systems of CS and its derivatives, we will encapsulate diclofenac sodium using three cross-
restrict our discussions only on these aspects. linking agents viz, glutaraldehyde, sulfuric acid and
heat treatment. Microspheres were spherical with
2.1. Emulsion cross-linking smooth surfaces as shown in Fig. 3. The size of the
microparticles ranged between 40 and 230 Am.
This method utilizes the reactive functional amine Among the three cross-linking agents used, glutaral-
group of CS to cross-link with aldehyde groups of the dehyde cross-linked microspheres showed the slowest
cross-linking agent (see Fig. 1b). In this method, a release rates while a quick release of diclofenac
water-in-oil (w/o) emulsion is prepared by emulsify- sodium was observed by the heat cross-linked micro-
ing the CS aqueous solution in the oil phase. Aqueous spheres. In our continuing study on CS-based
droplets are stabilized using a suitable surfactant. The derivatives [34], we have also prepared the nifedi-
stable emulsion is cross-linked by using an appro- pine-loaded microspheres of polyacrylamide-g-chito-
priate cross-linking agent such as glutaraldehyde to san using three concentrations of glutaraldehyde as
harden the droplets. Microspheres are filtered and the cross-linking agent. Microspheres were spherical
washed repeatedly with n-hexane followed by alcohol with the mean particle size of 450 Am.
and then dried [44]. By this method, size of the Glutaraldehyde extracted in toluene was used as a
particles can be controlled by controlling the size of cross-linking agent by Al-Helw et al. [45] to prepare
aqueous droplets. However, the particle size of final CS microspheres encapsulated with phenobarbitone.
product depends upon the extent of cross-linking Uniform and spherical microspheres with loading
agent used while hardening in addition to speed of efficiency up to 57.2% were produced. Loading
stirring during the formation of emulsion. This efficiency was dependent upon the preparation
method is schematically represented in Fig. 2. The conditions. Parameters affecting the preparation and
emulsion cross-linking method has few drawbacks performance of microspheres are molecular weight
S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28 9

spheres for intranasal delivery. Formulation parame-


ters such as drug loading, polymer concentration,
stirring speed during cross-linking and oil phase were
altered to develop microspheres having good in vivo
performance. In vivo studies indicated a significantly
improved bioavailability of pentazocine. Application
of in vitro data to various kinetic models indicated that
these systems followed the diffusion controlled
release kinetics.
Jameela et al. [48] prepared smooth, highly
spherical, cross-linked CS microspheres in the size
range of 45–300 Am for the controlled release (CR) of
progesterone. An aqueous acetic acid dispersion of CS
containing progesterone was emulsified in the dis-
persion medium consisting of liquid paraffin and
Fig. 3. Scanning electron micrograph of chitosan microspheres petroleum ether stabilized by using sorbitan sesquio-
produced by emulsion cross-linking method. leate; droplets were hardened by glutaraldehyde cross-
linking. Extent of cross-linking showed a significant
and concentration of CS as well as concentration of influence on drug release characteristics. Highly
the stabilizing agent. Particle size of the microspheres cross-linked microspheres released only about 35%
varied in the range 274–450 Am. Release rates of of steroid in 40 days compared to 70% release from
phenobarbitone from different formulations of micro- the lightly cross-linked microspheres. Evaluation of in
spheres showed high initial release (burst effect) of vivo bioavailability by intramuscular injection in
the drug and about 20–30% of the drug was released rabbits showed that a plasma concentration of 1 to 2
in the first hour. Release was faster from the small ng/mL was maintained up to 5 months without
size microspheres, i.e., almost 75–95% of the drug showing any high burst release effect. These
was released within 3 h depending upon the data suggest the usefulness of cross-linked CS
molecular weight of CS. Denkbas et al. [46] used microspheres as potential carriers for long-term
the mixture of mineral oil/petroleum ether in the ratio delivery of steroids. Bugamelli et al. [49] developed
of 60/40 (v/v) as the external medium to prepare CS insulin-loaded microparticles of CS by the interfacial
microspheres using glutaraldehyde as a cross-linking cross-linking in the presence of ascorbyl palmitate.
agent and Tween-80 as an emulsifier. Smaller micro- Disposition of ascorbyl palmitate at the water– oil
spheres with narrow distributions were produced interface allowed the formation of covalent bond with
when CS/solvent ratio and drug/CS ratio were lower. the amino groups of CS when its oxidation to
The 5-fluorouracil was loaded up to a concentration dehydroascorbyl palmitate took place during the
of 10.4 mg/g of CS. formation of microparticles. This method produced
Thanoo et al. [29] prepared the CS microspheres microparticles with high loading efficiency and
by emulsion cross-linking of CS solution in paraffin released the drug at a constant rate up to 80 h.
oil as an external medium with glutaraldehyde using
dioctyl sulfosuccinate as the stabilizing agent. Addi- 2.2. Coacervation/precipitation
tion of stabilizing agent during particle formation
produced microspheres with spherical geometry and This method utilizes the physicochemical property
smooth surfaces. Encapsulation efficiencies up to 80% of CS since it is insoluble in alkaline pH medium, but
were achieved for theophylline, aspirin or griseoful- precipitates/coacervates when it comes in contact with
vin. These microspheres were used to study the drug alkaline solution. Particles are produced by blowing
release rates, which were influenced by cross-link CS solution into an alkali solution like sodium
density, particle size and initial drug loading. Sankar hydroxide, NaOH-methanol or ethanediamine using
et al. [47] prepared the CS-based pentazocine micro- a compressed air nozzle to form coacervate droplets
10 S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28

[50]. Separation and purification of particles was done Chitosan–DNA nanoparticles have been prepared
by filtration/centrifugation followed by successive using the complex coacervation technique [53].
washing with hot and cold water. The method is Important parameters such as concentrations of
schematically represented in Fig. 4. Varying com- DNA, CS, sodium sulfate, temperature, pH of the
pressed air pressure or spray-nozzle diameter con- buffer and molecular weights of CS and DNA have
trolled the size of the particles and then using a cross- been investigated. At the amino to phosphate group
linking agent to harden particles can control the drug ratio between 3 and 8 and CS concentration of 100 Ag/
release. In another technique [51], sodium sulfate mL, the particle size was optimized to 100–250 nm
solution was added dropwise to an aqueous acidic with a narrow distribution. Surface charge of these
solution of CS containing a surfactant under stirring particles was slightly positive with a zeta potential of
and ultrasonication for 30 min. Microspheres were 112 to 118 mV at pH lower than 6.0, and became
purified by centrifugation and re-suspended in demin- nearly neutral at pH 7.2. The chitosan–DNA nano-
eralized water. Particles were cross-linked with particles could partially protect the encapsulated
glutaraldehyde. Particles produced by this method plasmid DNA from nuclease degradation.
have better acid stability than observed by other
methods. 2.3. Spray-drying
Chitosan microspheres loaded with recombinant
human interleukin-2 (rIL-2) have been prepared by Spray-drying is a well-known technique to pro-
dropping of rIL-2 with sodium sulfate solution in duce powders, granules or agglomerates from the
acidic CS solution [52]. When protein and sodium mixture of drug and excipient solutions as well as
sulfate solutions were added to CS solution and suspensions. The method is based on drying of
during the precipitation of CS, the protein was atomized droplets in a stream of hot air. In this
incorporated into microspheres. This method is method, CS is first dissolved in aqueous acetic acid
devoid of cross-linking agent. The rIL-2 was released solution, drug is then dissolved or dispersed in the
from microspheres in a sustained manner for up to 3 solution and then, a suitable cross-linking agent is
months. Efficacy of the systems developed was added. This solution or dispersion is then atomized
studied by using two model cells viz., HeLa and L- in a stream of hot air. Atomization leads to the
strain cell lines. Microspheres were taken up by the formation of small droplets, from which solvent
cells and rIL-2 was released from the microspheres. evaporates instantaneously leading to the formation
of free flowing particles [54] as depicted in Fig. 5.
Various process parameters are to be controlled to
get the desired size of particles. Particle size depends
upon the size of nozzle, spray flow rate, atomization
pressure, inlet air temperature and extent of cross-
linking.
He et al. [54] prepared both un-cross-linked and
cross-linked CS microparticles by spray-drying
method for the delivery of cimetidine, famotidine
and nizatidine. Microspheres were spherical with a
smooth and distorted morphology. Particle size of the
un-cross-linked microspheres varied between 4 and 5
Am, while cross-linked microspheres ranged from 2 to
10 Am; they were all positively charged. Particle size
and zeta potential were influenced by the extent of
cross-linking. A decrease in extent of cross-linking
increased both the particle size and the zeta potential.
Fig. 4. Schematic representation of preparation of chitosan Particle size was increased when the spray flow rate
particulate systems by coacervation/precipitation method. was increased using the large size nozzle. Micro-
S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28 11

Fig. 5. Schematic representation of preparation of chitosan particulate systems by spray drying method.

spheres with smaller particle size were produced at spherical in shape with smooth surfaces and narrow-
greater airflow rates. However, particle size was less size distributions.
affected by the inlet air temperature between 140 and Lorenzo-Lamosa et al. [60] prepared the micro-
180 8C. Conti et al. [55] produced microparticles by encapsulated CS microspheres for colonic delivery of
exposing the spray-dried particles to vapors contain- sodium diclofenac. Sodium diclofenac was entrapped
ing cross-linking agents. Cetylpyridinium chloride, an into CS microcores by spray-drying and then, micro-
anti-infective agent, was incorporated into CS micro- encapsulated into EudragitR L-100 and EudragitR S-
spheres produced by spray-drying technique. Extent 100 using an oil-in-oil solvent evaporation method.
of cross-linking was controlled by the time of By spray-drying, CS microspheres of 1.8–2.9 Am
exposure to cross-linking agent. sizes were prepared and efficiently microencapsulated
Ganza-Gonzalez et al. [56] have demonstrated that into EudragitR microspheres ranging in size between
spray-drying technique is fast, simple and reliable to 152 and 223 Am to form the multireservoir system.
obtain microspheres. Microspheres were prepared by Number of variables such as type and concentration of
spray drying of aqueous CS dispersions containing chitosan, the core/coat ratio and the type of enteric
metoclopramide hydrochloride using different polymer have been investigated to optimize the
amounts of formaldehyde as a cross-linker. Micro- microsphere properties. Huang et al. [61] prepared
spheres released the drug for more than 8 h, CS microspheres by the spray-drying method using
independent of the pH of the medium. In another type-A gelatin and ethylene oxide– propylene oxide
study [57], vitamin D2 (VD2), also called as ergo- block copolymer as modifiers. Surface morphology
calciferol, was efficiently encapsulated into CS micro- and surface charges of the prepared microspheres
spheres prepared by spray-drying method. The were investigated using SEM and microelectropho-
microencapsulated product was coated with ethyl resis. Shape, size and surface morphology of the
cellulose. The sustained release property of VD2 microspheres were significantly influenced by the
microspheres was used for the treatment of prostatic concentration of gelatin. Betamethasone disodium
disease [58]. Spray-drying method was also used to phosphate-loaded microspheres demonstrated a good
prepare ampicillin-loaded methylpyrrolidone CS drug stability (less 1% hydrolysis product), high
microspheres [59] by taking different drug-to-polymer entrapment efficiency (95%) and positive surface
weight ratios. Spray-dried microparticles were almost charge (37.5 mV). In vitro drug release from the
12 S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28

microspheres was related to gelatin content. Micro- prepared by this method for gadolinium neutron-
spheres containing gelatin/CS ratio of 0.4–0.6 (w/w) capture therapy. Particle size depends upon the type of
showed a prolonged release up to 12 h. CS, i.e., as the % deacetylation degree of CS
decreased, particle size increased, but drug content
2.4. Emulsion-droplet coalescence method decreased. Particles produced using 100% deacety-
lated CS had the mean particle size of 452 nm with
The novel emulsion-droplet coalescence method 45% drug loading. Nanoparticles were obtained
was developed by Tokumitsu et al. [62], which within the emulsion-droplet. Size of the nanoparticle
utilizes the principles of both emulsion cross-linking did not reflect the droplet size. Since gadopentetic
and precipitation. However, in this method, instead of acid is a bivalent anionic compound, it interacts
cross-linking the stable droplets, precipitation is electrostatically with the amino groups of CS, which
induced by allowing coalescence of CS droplets with would not have occurred if a cross-linking agent is
NaOH droplets. First, a stable emulsion containing used that blocks the free amino groups of CS. Thus, it
aqueous solution of CS along with drug is produced in was possible to achieve higher gadopentetic acid
liquid paraffin oil and then, another stable emulsion loading by using the emulsion-droplet coalescence
containing CS aqueous solution of NaOH is produced method compared to the simple emulsion cross-
in the same manner. When both emulsions are mixed linking method.
under high-speed stirring, droplets of each emulsion
would collide at random and coalesce, thereby 2.5. Ionic gelation
precipitating CS droplets to give small size particles.
The method is schematically shown in Fig. 6. The use of complexation between oppositely
Gadopentetic acid-loaded CS nanoparticles have been charged macromolecules to prepare CS microspheres
has attracted much attention because the process is
very simple and mild [63,64]. In addition, reversible
physical cross-linking by electrostatic interaction,
instead of chemical cross-linking, has been applied
to avoid the possible toxicity of reagents and other
undesirable effects. Tripolyphosphate (TPP) is a
polyanion, which can interact with the cationic CS
by electrostatic forces [65,66]. After Bodmeier et al.
[67] reported the preparation of TPP–CS complex by
dropping CS droplets into a TPP solution, many
researchers have explored its potential pharmaceutical
usage [68–73]. In the ionic gelation method, CS is
dissolved in aqueous acidic solution to obtain the
cation of CS. This solution is then added dropwise
under constant stirring to polyanionic TPP solution.
Due to the complexation between oppositely charged
species, CS undergoes ionic gelation and precipitates
to form spherical particles. The method is schemati-
cally represented in Fig. 7. However, TPP/CS micro-
particles formed have poor mechanical strength thus,
limiting their usage in drug delivery.
Insulin-loaded CS nanoparticles have been pre-
pared by mixing insulin with TPP solution and then
adding this to CS solution under constant stirring [74].
Fig. 6. Schematic representation of preparation of chitosan Two types of CS in the form of hydrochloride salt
particulate systems by emulsion-droplet coalescence method. (SeacureR 210 Cl and ProtasanR 110 Cl), varying in
S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28 13

Fig. 7. Schematic representation of preparation of chitosan particulate systems by ionic gelation method.

their molecular weight and degree of deacetylation, the aqueous solution of CS in vivo. After adminis-
were utilized for nanoparticle preparation. For both tration of 21 I.U./kg insulin in the CS nanoparticles,
types of CS, TPP concentration was adjusted to get a hypoglycemia was prolonged over 15 h. The average
CS/TPP ratio of 6:1. Chitosan nanoparticles thus pharmacological bioavailability relative to s.c. injec-
obtained were in the size range of 300–400 nm with a tion of insulin solution was up to 14.9%.
positive surface charge ranging from +54 to +25 mV. Xu and Du [76] have studied different formulations
Using this method, insulin loading was modulated of CS nanoparticles produced by the ionic gelation of
reaching the values up to 55%. Efficiency of the TPP and CS. TEM indicated their diameter ranging
method was dependent upon the deacetylation of CS, between 20 and 200 nm with spherical shape. FTIR
since it involves the gelation of protonated amino confirmed tripolyphosphoric groups of TPP linked
groups of CS. with ammonium groups of CS in the nanoparticles.
There are many ongoing investigations, which Factors that affect the delivery of bovine serum
demonstrate the improved oral bioavailability of albumin (BSA) as a model protein have been studied.
peptide and protein formulations. Bioadhesive poly- These include molecular weight and deacetylation
saccharide CS nanoparticles would seem to further degree of CS, concentrations of CS and BSA, as well
enhance their intestinal absorption. Pan et al. [75] as the presence of polyethylene glycol (PEG) in the
prepared the insulin-loaded CS nanoparticles by encapsulation medium. Increasing molecular weight
ionotropic gelation of CS with TPP anions. Particle of CS from 10 to 210 kDa, BSA encapsulation
size distribution and zeta potential were determined by efficiency was enhanced nearly twice. The total
photon correlation spectroscopy. The ability of CS release of BSA in phosphate buffered saline pH 7.4
nanoparticles to enhance the intestinal absorption of in 8 days was reduced from 73.9% to 17.6%.
insulin and the relative pharmacological bioavailability Increasing deacetylation degree from 75.5% to 92%
of insulin was investigated by monitoring the plasma promoted the encapsulation efficiency with a decrease
glucose level of alloxan-induced diabetic rats after the in release rate. Encapsulation efficiency decreased
oral administration of various doses of insulin-loaded greatly by increasing the initial concentration of BSA
CS nanoparticles. The positively charged, stable CS and CS. Higher loading capacity of BSA enhanced the
nanoparticles showed particle size in the range of 250– BSA release from nanoparticles. However, adding
400 nm. Insulin association was up to 80%. The in vitro PEG hindered the BSA encapsulation and increased
release experiments indicated initial burst effect, which the release rate.
is pH-sensitive. The CS nanoparticles enhanced the Ko et al. [77] prepared CS microparticles with TPP
intestinal absorption of insulin to a greater extent than by the ionic cross-linking method. Particle sizes of
14 S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28

TPP-CS microparticles varied from 500 to 710 Am [79], and these droplets are highly monodispersed,
with drug encapsulation efficiencies more than 90%. preparation of drug-loaded nanoparticles in reverse
Morphologies of TPP-CS microparticles have been micelles will produce extremely fine particles with a
examined by SEM. As the pH of TPP solution narrow size distribution. Since micellar droplets are
decreased and molecular weight of CS increased, in Brownian motion, they undergo continuous
microparticles acquired better spherical shape having coalescence followed by re-separation on a time-
smooth surface. Release of felodipine as a model drug scale that varies between millisecond and micro-
was affected by the preparation method. Chitosan second [80]. The size, polydispersity and thermody-
microparticles prepared at lower pH or higher namic stability of these droplets are maintained in the
concentration of TPP solution resulted in a slower system by a rapid dynamic equilibrium.
release of felodipine. With a decreasing molecular In this method, the surfactant is dissolved in a
weight and concentration of CS solution, the drug organic solvent to prepare reverse micelles. To this,
release increased. The release of drug from TPP-CS aqueous solutions of CS and drug are added with
microparticles decreased when the cross-linking time constant vortexing to avoid any turbidity. The aqueous
was increased. phase is regulated in such a way as to keep the entire
mixture in an optically transparent microemulsion
2.6. Reverse micellar method phase. Additional amount of water may be added to
obtain nanoparticles of larger size. To this transparent
Reverse micelles are thermodynamically stable solution, a cross-linking agent is added with constant
liquid mixtures of water, oil and surfactant. Macro- stirring, and cross-linking is achieved by stirring
scopically, they are homogeneous and isotropic, overnight. The maximum amount of drug that can
structured on a microscopic scale into aqueous and be dissolved in reverse micelles varies from drug to
oil microdomains separated by surfactant-rich films. drug and has to be determined by gradually increasing
One of the most important aspects of reverse micelle the amount of drug until the clear microemulsion is
hosted systems is their dynamic behavior. Nano- transformed into a translucent solution. The organic
particles prepared by conventional emulsion poly- solvent is then evaporated to obtain the transparent
merization methods are not only large (N200 nm), dry mass. The material is dispersed in water and then
but also have a broad size range. Preparation of adding a suitable salt precipitates the surfactant out.
ultrafine polymeric nanoparticles with narrow size The mixture is then subjected to centrifugation. The
distribution could be achieved by using reverse supernatant solution is decanted, which contains the
micellar medium [78]. Aqueous core of the reverse drug-loaded nanoparticles. The aqueous dispersion is
micellar droplets can be used as a nanoreactor to immediately dialyzed through dialysis membrane for
prepare such particles. Since the size of the reverse about 1 h and the liquid is lyophilized to dry powder.
micellar droplets usually lies between 1 and 10 nm The method is schematically represented in Fig. 8.

Fig. 8. Schematic representation of preparation of chitosan particulate systems by reverse micellar method.
S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28 15

Mitra et al. [81] have encapsulated doxorubicin– jelly mass that was cross-linked by adding glutaralde-
dextran conjugate in CS nanoparticles prepared by hyde. The non-sticky cross-linked mass was passed
reverse micellar method. The surfactant sodium through a sieve with a suitable mesh size to get
bis(ethyl hexyl) sulfosuccinate (AOT), was dissolved microparticles. The microparticles were washed with
in n-hexane. To 40 mL of AOT solution (0.03 M), 100 0.1 N NaOH solution to remove the un-reacted excess
AL of 0.1% CS solution in acetic acid, 200 AL glutaraldehyde and dried overnight in an oven at 40
doxorubicin–dextran conjugate (6.6 mg/mL), 10 AL 8C. Clozapine was incorporated into CS before cross-
liquor ammonia and 10 AL of 0.01% glutaraldehyde linking with an entrapment efficiency up to 98.9%.
solution were added with continuous stirring at room This method is devoid of tedious procedures, and can
temperature. This procedure produced CS nanopar- be scaled up easily. Microparticles were irregular in
ticles encapsulating doxorubicin–dextran conjugate. shape, with the average particle sizes in the range
Solvent was removed by rotary evaporator and the dry 543–698 Am. The in vitro release was extended up to
mass was resuspended in 5 mL of pH 7.4 Tris–Cl 12 h, while the in vivo studies indicated a slow release
buffer by sonication. To this, 1 mL of 30% CaCl2 of clozapine.
solution was added dropwise to precipitate the
surfactant as calcium salt of diethylhexyl sulfosucci-
nate. The precipitate was pelleted by centrifugation at 3. Drug loading into micro/nanoparticles of
5,000 rpm for 30 min at 4 8C. The pellet was chitosan
discarded and the supernatant containing nanopar-
ticles was centrifuged at 60,000 rpm for 2 h to pellet Drug loading in micro/nanoparticulate systems can
the nanoparticles. The pellet was dispersed in 5 mL of be done by two methods, i.e., during the preparation
pH 7.4 Tris–HCl buffer. of particles (incorporation) and after the formation of
particles (incubation). In these systems, drug is
2.7. Sieving method physically embedded into the matrix or adsorbed onto
the surface. Various methods of loading have been
Recently, Agnihotri and Aminabhavi [82] have developed to improve the efficiency of loading, which
developed a simple, yet novel method to produce CS largely depends upon the method of preparation as
microparticles. In this method, microparticles were well as physicochemical properties of the drug.
prepared by cross-linking CS to obtain a non-sticky Maximum drug loading can be achieved by incorpo-
glassy hydrogel followed by passing through a sieve rating the drug during the formation of particles, but it
as shown in Fig. 9. A suitable quantity of CS was may get affected by the process parameters such as
dissolved in 4% acetic acid solution to form a thick method of preparation, presence of additives, etc.

Fig. 9. Schematic representation of preparation of chitosan particulate systems by sieving method.


16 S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28

Both water-soluble and water-insoluble drugs can mixed with CS solution before simultaneous cross–
be loaded into CS-based particulate systems. Water- linking and precipitation. In method II, drug was
soluble drugs are mixed with CS solution to form a incubated with the pre-formed microspheres for 48
homogeneous mixture, and then, particles can be h. Cumulative amount of tetracycline that was
produced by any of the methods discussed before. released from CS microspheres and stability of drug
For instance, cisplatin was loaded [83] during the was examined in different pH media at 37 8C.
formation of particles with encapsulation efficiency Microspheres with a spherical shape having an
as high as 99%. The initial concentration of average diameter of 2– 3 Am were formed. When
cisplatin and volume of glutaraldehyde had no drug was added to CS solution before cross-linking
effect on the encapsulation efficiency. Drug encap- and precipitation, only 8% (w/w) was optimally
sulation increased as the concentration of CS incorporated in the final microsphere formulation.
increased. Water-insoluble drugs and drugs that When drug was incubated with the pre-formed
can precipitate in acidic pH solutions can be loaded microspheres, a maximum of 69% (w/w) could be
after the formation of particles by soaking the loaded. About 30% of tetracycline either in solution
preformed particles with the saturated solution of or when released from the microspheres was found
drug. to degrade at pH 1.2 in 12 h. Preliminary results of
Diclofenac sodium, which precipitates in acidic pH this study suggested that CS microspheres can be
conditions, has been loaded by the soaking method used to incorporate antibiotic drugs, which may be
[33]. In this method, loading depends upon the effective when administered locally in the stomach
swelling of particles in water. Percentage loading of against H. pylori.
drug decreased with increasing cross-linking due to
decreased swelling. Water-insoluble drugs can also be
loaded using the multiple emulsion technique. In this 4. Drug release and release kinetics
method, drug is dissolved in a suitable solvent and
then emulsified in CS solution to form an oil-in-water Drug release from CS-based particulate systems
(o/w) type emulsion. Sometimes, drug can be dis- depends upon the extent of cross– linking, morphol-
persed into CS solution by using a surfactant to get the ogy, size and density of the particulate system,
suspension. Thus, prepared o/w emulsion or suspen- physicochemical properties of the drug as well as
sion can be further emulsified into liquid paraffin to the presence of adjuvants. In vitro release also
get the oil-water-oil (o/w/o) multiple emulsion. The depends upon pH, polarity and presence of enzymes
resulting droplets can be hardened by using a suitable in the dissolution media. The release of drug from CS
cross-linking agent. particulate systems involves three different mecha-
In a study by Jameela et al. [84], bovine serum nisms: (a) release from the surface of particles, (b)
albumin (BSA) and diphtheria toxoid were loaded diffusion through the swollen rubbery matrix and (c)
into preformed glutaraldehyde cross-linked CS release due to polymer erosion. These mechanisms are
microspheres by passive absorption from aqueous shown schematically in Fig. 10.
solutions. This method is an alternative to loading In majority of cases, drug release follows more
biological macromolecules that are sensitive to than one type of mechanism. In case of release
organic solvents, pH, temperature, ultrasound, etc. from the surface, adsorbed drug instantaneously
In vitro release of BSA showed a high burst effect. dissolves when it comes in contact with the release
Coating of particles with paraffin or polylactic acid medium. Drug entrapped in the surface layer of
modulated the drug release. Diphtheria toxoid loaded particles also follows this mechanism. This type of
CS microspheres showed constant antibody titres for drug release leads to burst effect. He et al. [54]
5 months. observed that cemetidine-loaded CS microspheres
Hejazi and Amiji [85] have prepared CS micro- have shown burst effect in the early stages of
spheres by ionic cross-linking and precipitation with dissolution. Most of the drug was released within
sodium sulfate. Two different methods were used few minutes when particles were prepared by spray
for drug loading. In method I, tetracycline was drying technique. Increasing the cross-linking den-
S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28 17

Fig. 10. Mechanism of drug release from particulate systems.

sity can prevent the burst release. This effect can within the size range of 250– 500 Am, but for
also be avoided by washing microparticles with a particles in the size range of 500– 1,000 Am, drug
proper solvent, but it may lead to low encapsulation release was 56– 90% in 5 h. This is attributed to
efficiency. large surface area available for dissolution with a
Drug release by diffusion involves three steps. small particle size, thus favoring rapid release of the
First, water penetrates into particulate system, which drug compared to larger microspheres.
causes swelling of the matrix; secondly, the Kweon and Kang [86] prepared the CS-g–
conversion of glassy polymer into rubbery matrix poly(vinyl alcohol) matrix to study the release of
takes place, while the third step is the diffusion of prednisolone under various conditions. Relationship
drug from the swollen rubbery matrix. Hence, the between the amount of drug release and square root
release is slow initially and later, it becomes fast. of time was linear indicating the diffusion-controlled
This type of release is more prominent in case of release. Drug release was controlled by the extent of
hydrogels. Al-Helw et al. [45] observed a high PVA grafting, heat treatment or cross-link density,
initial release of the drug in all the prepared but it was less affected by the pH when compared to
formulations. Nearly, 20– 30% of the incorporated plain chitosan. Ganza-Gonzalez et al. [56] analyzed
drug was released in the first hour. Release was the drug release data using Higuchi equation [87].
dependent on the molecular weight of CS and Higuchi equation was used to describe the release of
particle size of the microspheres. The release rate a solute from a flat surface, but not from a sphere
from microspheres prepared from high molecular [88], but the good fit obtained suggests that the
weight CS was slow compared to those prepared release rate depends upon the rate of diffusion
from medium and low molecular weight CS. This through the cross-linked matrix. Authors have also
could be attributed to both lower solubility of high fitted the release data to equations developed by Guy
molecular weight CS and higher viscosity of the gel et al. [89] to describe the diffusion from a sphere.
layer formed around the drug particles upon contact The most commonly used equation for diffusion-
with the dissolution medium. The release within the controlled matrix system is an empirical equation used
first 3 h was fast (75– 95%) from microspheres by Ritger and Peppas [90], in which the early time
18 S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28

release data can be fitted to obtain the diffusion Higuchi equation [87]. It was demonstrated that the
parameters, rate of release depends upon the size of microspheres.
Release from smaller size microspheres was faster
Mt
¼ kt n ð1Þ than those from the large size microspheres due to
Ml
smaller diffusional path length for the drug and the
Here, M t /M l is the fractional drug release at time t, k larger surface area of contact of smaller particles with
is a constant characteristic of the drug-polymer the dissolution medium. Orienti et al. [92] studied the
interaction and n is an empirical parameter character- correlation between matrix erosion and release
izing the release mechanism. Based on the diffusional kinetics of indomethacin-loaded CS microspheres.
exponent [91], drug transport is classified as Fickian Release kinetics was correlated with the concentration
(n=0.5), Case II transport (n=1), non-Fickian or of CS in the microsphere and pH of the release
anomalous (0.5bnb1) and super Case II (nN1). Drug medium. At high concentrations of CS and at pH 7.4,
release from the CS microspheres cross– linked with deviations from Fickian to zero order kinetics have
glutaraldehyde, sulfuric acid and heat have shown been observed. Variations induced by these parame-
[33] different n values varying from 0.47 to 0.61. The ters on drug diffusion and solubility in the matrix
n values increase with increasing loading of diclofe- undergoing erosion have been analyzed.
nac sodium in different cross-linked formulations.
Recently, Agnihotri and Aminabhavi [82] analyzed
the dynamic swelling data of CS microparticles using 5. Pharmaceutical applications of chitosan
Eq. (1) to predict drug release from the water uptake particulate systems
data of the microparticles cross-linked with (5.0, 7.5
and 10.0)10– 4 mL of glutaraldehyde/mg of CS. It Chitosan-based particulate systems are attracting
was observed that as the cross-linking increases, pharmaceutical and biomedical applications as poten-
swelling of CS microparticles decreases. Values of n tial drug delivery devices. Some important applica-
obtained in the range of 0.160 to 0.249 indicating that tions are discussed below.
the release mechanism deviates from the Fickian
trend. The values of n are b0.5 due to the irregular 5.1. Colon targeted drug delivery
shaped particles and these decrease systematically
with increasing cross-linking. Chitosan is a promising polymer for colon drug
In the swelling controlled release systems, drug is delivery since it can be biodegraded by the colonic
dispersed within a glassy polymer. Upon contact with bacterial flora [93,94] and it has mucoadhesive
biological fluid, the polymer swells, but no drug character [1]. The pH-sensitive multicore microparti-
diffusion occurs through the polymer phase. As the culate system containing CS microcores entrapped
penetrant enters the glassy polymer, glass transition into enteric acrylic microspheres was reported [60].
temperature of the polymer is lowered due to Sodium diclofenac was efficiently entrapped within
relaxation of the polymer chains. Drug could diffuse these CS microcores and then microencapsulated into
out of the swollen rubbery polymer. This type of Eudragit L-100 and Eudragit S-100 to form a multi-
system is characterized by two moving boundaries: reservoir system. In vitro release study revealed no
the front separating the swollen rubbery portion and release of the drug in gastric pH for 3 h and after the
the glassy region, which moves with a front velocity lag-time, a continuous release for 8– 12 h was
and the polymer fluid interface. The rate of drug observed in the basic pH.
release is controlled by the velocity and position of the
front dividing the glassy and rubbery portions of the 5.2. Mucosal delivery
polymer.
Jameela et al. [48] have obtained a good correlation Nowadays, mucosal surfaces such as nasal, peroral
fit for the cumulative drug released vs. square root of and pulmonary are receiving a great deal of attention
time, demonstrating that the release from the micro- as alternative routes of systemic administration.
sphere matrix is diffusion-controlled and obeys Chitosan has mucoadhesive properties and therefore,
S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28 19

it seems particularly useful to formulate the bioadhe- (CLSM). Chitosan microparticles were prepared using
sive dosage forms for mucosal administration (ocular, a precipitation/coacervation method. The size of CS
nasal, buccal, gastro-enteric and vaginal-uterine ther- microparticles was 4.3±±0.7 Am and were positively
apy) [95]. Nasal mucosa has high permeability and charged (20±1 mV). Since only microparticles smaller
easy access of drug to the absorption site. The than 10 Am can be taken up by M-cells of Peyer’s
particulate delivery to peroral mucosa is easily taken patches, these microparticles were used as vaccination
up by the Peyer’s patches of the gut associated systems. The CLSM studies showed that the model
lymphoid tissue. Chitosan has been found to enhance antigen ovalbumin was entrapped within the CS
the drug absorption through mucosae without damag- microparticles. Field emission scanning electron
ing the biological system. Here, the mechanism of microscopy demonstrated the porous structure of CS
action of CS was suggested to be a combination of microparticles, thus facilitating the entrapment of
bioadhesion and a transient widening of the tight ovalbumin. Ovalbumin loading in CS microparticles
junctions between epithelial cells [27]. was about 40%. Release studies have shown the low
Genta et al. [95] studied the influence of gluta- release of ovalbumin within 4 h, but most of
raldehyde on drug release and mucoadhesive proper- ovalbumin (about 90%) remained entrapped in the
ties of CS microspheres. A new in vitro technique was microparticles. Since CS microparticles are biode-
developed based on electron microscopy to study the gradable, the entrapped ovalbumin was released after
effect of polymer cross-link density on the mucoad- intracellular digestion in Peyer’s patches. Initial in
hesive properties of CS microspheres modulating the vivo studies demonstrated that fluorescently labeled
rate of theophylline release. The ability of insulin- CS microparticles can be taken up by the epithelium
loaded CS nanoparticles to enhance the nasal absorp- of the murine Peyer’s patches. Since the uptake by
tion of insulin was investigated in a conscious rabbit Peyer’s patches is an essential step in oral vaccination,
model. Chitosan nanoparticles enhanced the nasal these results have shown that the porous CS micro-
absorption of insulin to a greater extent than the particles developed are most promising vaccine
aqueous solution of CS [74]. van der Lubben et al. delivery systems.
[96] incorporated the model protein ovalbumin into
CS microparticles and the uptake of ovalbumin 5.3. Cancer therapy
associated with CS microparticles in murine Peyer’s
patches was demonstrated using confocal laser scan- Gadopentetic acid-loaded CS nanoparticles have
ning microscopy. In a further study, van der Lubben et been prepared for gadolinium neutron-capture therapy
al. [97] investigated the ability of CS microparticles to [62]. Their releasing properties and ability for long-
enhance both systemic and local immune responses term retention of gadopentetic acid in the tumor
against diphtheria toxoid (DT) vaccine after the oral indicated that these nanoparticles are useful as intra-
and nasal administration in mice. Systemic and local tumoral injectable devices for gadolinium neutron-
IgG and IgA immune responses against DT associated capture therapy. The accumulation of gadolinium
to CS microparticles were strongly enhanced after the loaded as gadopentetic acid (Gd-DTPA) in CS nano-
oral delivery in mice. particles designed for gadolinium neutron-capture
Even though oral vaccination has numerous therapy (Gd-NCT) for cancer have been evaluated in
advantages over the parenteral injection, degradation vitro in cultured cells [99]. Using L929 fibroblast
of the vaccine in the gut and low uptake in the cells, Gd accumulation for 12 h at 37 8C was
lymphoid tissue of the gastrointestinal tract still investigated at Gd concentrations lower than 40
complicate the development of oral vaccines. In this ppm. The accumulation leveled above 20 ppm and
direction, van der Lubben et al. [98] prepared the CS reached 18.0±2.7 (mean±S.D.) Ag Gd/106 cells at 40
microparticles and characterized them for size, zeta ppm. Furthermore, the corresponding accumulations
potential, morphology- and ovalbumin-loading as in B16F10 melanoma cells and SCC-VII squamous
well as release characteristics. The in vivo uptake of cell carcinoma, which were used in the previous Gd-
CS microparticles by murine Peyer’s patches was NCT trials in vivo were 27.1±2.9 and 59.8±9.8 Ag Gd/
studied by using confocal laser scanning microscopy 106 cells, respectively. This explains the superior
20 S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28

growth-suppression in the in vivo trials using SCC- evaluated in J774A.1 macrophage tumor cells
VII cells. The accumulation of nanoparticles in these implanted in Balb/c mice. The in vivo efficacy of
cells was 100– 200 times higher in comparison to these nanoparticles was determined by tumor regres-
dimeglumine gadopentetate aqueous solution (Mag- sion and increased survival time compared to doxor-
nevistw), a magnetic resonance imaging contrast ubicin– dextran conjugate and the free drug. These
agent. The endocytic uptake of nanoparticles was results suggest that the system not only reduced the
suggested from TEM. These findings indicated that side effects, but also improved its therapeutic efficacy
nanoparticles had a high affinity to cells, thus in the treatment of solid tumors.
contributing to the long retention of Gd in tumor Janes et al. [103] evaluated the potential of CS
tissue leading to significant suppression of tumor nanoparticles as carriers for doxorubicin (DOX). The
growth in in vivo studies. challenge was to entrap a cationic, hydrophilic
Tokumitsu et al. [100] demonstrated the potential molecule into nanoparticles formed by ionic gelation
usefulness of Gd-NCT using gadolinium-loaded nano- of the positively charged CS. To achieve this
particles. The potential of gadolinium neutron-capture objective, the authors have masked the positive charge
therapy (Gd-NCT) for cancer was evaluated using CS of DOX by complexing it with dextran sulfate. This
nanoparticles as a novel gadolinium device. The modification doubled the DOX encapsulation effi-
nanoparticles incorporated with 1200 mg of natural ciency relative to controls and enabled real loadings
gadolinium were administered intratumorally twice in up to 4.0 wt.% of DOX. Authors also investigated the
mice-bearing subcutaneous B16F10 melanoma. The possibility of forming a complex between CS and
thermal neutron irradiation was performed for the DOX prior to the formation of particles. Despite low
tumor site, with the fluence of 6.321012 neutrons/ complexation efficiency, no dissociation of the com-
cm2, 8 h after the second gadolinium administration. plex was observed upon the formation of nano-
After irradiation, the tumor growth in the nano- particles. Fluorimetric analysis of the in vitro drug
particle-administered group was significantly sup- released showed the initial release phase, the intensity
pressed compared to that in the gadopentetate of which was dependent upon the association mode,
solution-administered group, despite radioresistance followed by a very slow release. Evaluation of the
of melanoma and the smaller Gd dose than that activity of DOX-loaded nanoparticles in cell cultures
administered in past Gd-NCT trials. indicated that those containing dextran sulfate were
Jameela et al. [101] have prepared glutaraldehyde able to maintain cytostatic activity relative to free
cross-linked CS microspheres containing mitoxan- DOX, while DOX complexed with CS before the
trone. The antitumor activity was evaluated against nanoparticle formation showed a slightly decreased
Ehrlich ascites carcinoma in mice by intraperitoneal activity. Additionally, confocal studies showed that
injections. The tumor inhibitory effect was followed DOX was not released in the cell culture medium, but
by monitoring the survival time and change in the entered the cells while being associated to nano-
body weight of the animal for 60 days. Mean survival particles. These studies have shown the feasibility of
time of animals which received free mitoxantrone was CS nanoparticles to entrap DOX and to deliver it to
2.1 days and this was increased to 50 days when the cells in its active form.
mitoxantrone was given via microspheres. In another
study [102], the in vitro release of mitoxantrone was 5.4. Gene delivery
controlled for 4 weeks in phosphate buffer at 27 8C.
Mitra et al. [81] have encapsulated doxorubicin– Gene therapy is a challenging task in the treatment
dextran conjugate into long circulating CS nano- of genetic disorders. In case of gene delivery, the
particles. In an attempt to minimize cardiotoxicity of plasmid DNA has to be introduced into the target
doxorubicin, a conjugate with dextran was prepared cells, which should get transcribed and the genetic
and encapsulated in CS nanoparticles. Size of the information should ultimately be translated into the
nanoparticle was 100±10 nm, which favors enhanced corresponding protein. To achieve this goal, number
permeability and retention effect. Antitumor effect of of hurdles are to be overcome by the gene delivery
these doxorubicin– dextran-loaded nanoparticles was system. Transfection is affected by: (a) targeting the
S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28 21

delivery system to target cell, (b) transport through the groups were chemically bound to CS for liver
cell membrane, (c) uptake and degradation in the specificity and dextran was grafted to increase the
endolysosomes and (d) intracellular trafficking of stability of the complex in water. It was shown that this
plasmid DNA to the nucleus. Chitosan could interact system could efficiently transfect liver cells.
ionically with the negatively charged DNA and forms Chew et al. [108] studied the i.m. immunization
polyelectrolyte complexes. In these complexes, DNA with full-length Der p 1 cDNA induced significant
becomes better protected against nuclease degradation humoral response to the left domain (approximately
leading to better transfection efficiency. corresponding to amino acids 1– 116), but not to the
DNA– CS nanoparticles have been prepared [53] right domain (approximately corresponding to amino
to examine the influence of several parameters on acids 117– 222) of Der p 1 allergen. Authors explored
their preparation. The transfection efficiency of CS- the use of CS– DNA nanoparticles for oral immuniza-
DNA nanoparticles was cell-type dependent. Typi- tion to induce the immune responses specific to both
cally, it was 3 to 4 orders of magnitude, in relative left and right domains of Der p 1. DNA constructs
light units, higher than the background level in pDer p 1 (1– 222) and pDer p 1 (114– 222), which
HEK293 cells, and 2 to 10 times lower than that were complexed with CS and delivered orally
achieved by LipofectAMINE–– DNA complexes. followed by an i.m. injection of pDer p 1 (1– 222)
The presence of 10% fetal bovine serum did not after 13 weeks. Such an approach has successfully
interfere with their transfection ability. The study primed Th1-skewed immune responses against both
also developed three different schemes to conjugate domains of Der p 1. It was suggested that such a
transferrin or KNOB protein to the nanoparticle strategy could be further optimized for more effica-
surface. The transferrin conjugation only yielded a cious gene vaccination for full-length Der p 1.
maximum of 4-fold increase in their transfection Numerous studies have been reported on prophy-
efficiency in HEK293 cells and HeLa cells, whereas lactic and therapeutic use of genetic vaccines for
KNOB conjugated nanoparticles could improve the combating a variety of infectious diseases in animal
gene expression level in HeLa cells by 130-fold. models. Recent human clinical studies with the gene
Conjugation of PEG on nanoparticles allowed gun have validated the concept of direct targeting of
lyophilization without aggregation, and without loss dendritic cells (Langerhan’s cells) in the viable
of bioactivity for at least 1 month in storage. The epidermis of the skin. However, it is unclear whether
clearance of PEGylated nanoparticles in mice follow- the gene gun technology or other needle-free devices
ing i.v. administration was slower than the unmodi- will become commercially viable. Cui and Mumper
fied nanoparticles at 15 min, and with higher [109] investigated the topical application of CS-
depositions in kidney and liver. However, no differ- based nanoparticles containing plasmid DNA
ence was observed during the first hour. (pDNA) as a potential approach to genetic immuni-
Self-aggregates were prepared [104] by hydro- zation. Two types of nanoparticles were investigated:
phobic modification of CS with deoxycholic acid in (i) pDNA-condensed CS nanoparticles and (ii)
aqueous media. Self-aggregates have a small size pDNA-coated on pre-formed cationic CS/carboxy-
(mean diameter of 160 nm) with an unimodal size methylcellulose (CMC) nanoparticles. These studies
distribution. Self-aggregates can form charge com- have shown that both CS and a CS oligomer can
plexes when mixed with plasmid DNA. The useful- complex CMC to form stable cationic nanoparticles
ness of self-aggregates/DNA complex for transfer of for subsequent pDNA coating. Selected pDNA-
genes into mammalian cells in vitro has been coated nanoparticles (with pDNA up to 400 mg/
suggested. Several transfection studies using chemi- mL) were stable to challenge with the serum.
cally modified CS have been reported. Trimethyl CS Several different CS-based nanoparticles containing
oligomers were examined for their potency as DNA pDNA resulted in both detectable and quantifiable
carriers [105]. Chitosan and lactosylated CS carriers levels of luciferase expression in mouse skin 24 h
were investigated for their transfection efficiencies in after topical application and significant antigen-
vitro [106]. Recently, galactosylated CS-g– dextran– specific IgG titer to expressed h-galactosidase at
DNA complexes have been prepared [107]. Galactose 28 days.
22 S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28

Borchard [110] has recently published a review on able CyA levels in the inner ocular structures (i.e., iris/
the efficient non-viral gene delivery using cationic ciliary body and aqueous humour), blood and plasma.
polymers as DNA-condensing agents. The gene These levels were significantly higher than those
delivery is dependent on several factors such as obtained following the instillation of CS solution
complex size, complex stability, toxicity, immunoge- containing CyA and an aqueous CyA suspension. The
nicity, protection against DNase degradation, intra- study indicated that CS nanoparticles could be used as
cellular trafficking and processing of the DNA. The a vehicle to enhance the therapeutic index of the
review also examined the advances made in the clinically challenging drugs with potential application
application of CS and CS derivatives to non-viral at the extraocular level.
gene delivery. It gives an overview of the transfection
studies performed by using CS as a transfection agent. 5.7. Chitosan as a coating material

5.5. Topical delivery Chitosan has good film forming properties and
hence, it is used as a coating material in drug delivery
Due to good bioadhesive property and ability to applications. Chitosan-coated microparticles have
sustain the release of the active constituents, CS has many advantages such as improvement of drug
been used in topical delivery systems. Bioadhesive CS payloads, bioadhesive property and prolonged drug
microspheres for topical sustained release of cetyl release properties over the uncoated particles. Chito-
pyridinium chloride have been evaluated [55]. san-coated microspheres composed of poly(lactic
Improved microbiological activity was shown by acid)– poly(caprolactone) blends have been prepared
these microparticulate systems. Conti et al. [111] [113]. These microspheres showed good potential for
prepared microparticles composed of CS and designed the targeted delivery of antiproliferative agents to treat
as powders for topical wound-healing properties. restenosis. Shu and Zhu [73] have prepared the
Blank and ampicillin-loaded microspheres were pre- alginate beads coated with CS by three different
pared by spray-drying technique. In vivo evaluation in methods. The release of brilliant blue was not only
albino rats showed that both drug-loaded and blank affected by CS density on the particle surface, but also
microspheres have shown good wound healing on the preparation method and other factors. Chiou et
properties. al. [114] have used different molecular weight
chitosans for coating the microspheres. The initial
5.6. Ocular delivery burst release was observed in the first hour with 50%
release of lidocaine. But, 19.2% release occurred at
De Campos et al. [112] investigated the potential of 25th hour for the un-coated particles and 14.6% at the
CS nanoparticles as a new vehicle to improve the 90th hour for the CS-coated microspheres.
delivery of drugs to ocular mucosa. Cyclosporin A
(CyA) was chosen as a model drug. A modified ionic
gelation technique was used to produce CyA-loaded 6. Chemically modified chitosans
CS nanoparticles. These nanoparticles with a mean
size of 293 nm, a zeta potential of +37 mV, high CyA Various chemical modifications of CS have been
association efficiency and loading of 73% and 9%, studied to alter its properties. N-Trimethyl chitosan
respectively were obtained. The in vitro release chloride (TMC), a quaternized CS derivative, has
studies, performed under sink conditions, revealed been proven to effectively increase the permeation of
the fast release during the first hour followed by a hydrophilic macromolecular drugs across- the
more gradual drug release during the 24-h period. The mucosal epithelia by opening the tight junctions
in vivo experiments showed that after topical instilla- [115]. The study investigated the intestinal absorption
tion of CyA-loaded CS nanoparticles to rabbits, of octreotide when it is co-administered with a
therapeutic concentrations were achieved in the polycationic absorption enhancer, TMC. Chitosan
external ocular tissues (i.e., cornea and conjunctiva) succinate and CS phthalate were synthesized and
within 48 h while maintaining negligible or undetect- assessed as potential matrices for colon-specific orally
S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28 23

administered drug delivery applications. The prepared fluorescence spectroscopy and dynamic light scatter-
matrices resisted the dissolution under acidic con- ing method. A charge complex was produced between
ditions. On the other hand, improved drug release the cationically charged self-aggregates and the
profiles were observed in basic conditions. These negatively charged plasmid DNA. The feasibility of
results suggested the suitability of the prepared self-aggregates as an in vitro delivery vehicle was
matrices in colon specific and orally administered investigated for the transfection of genetic material in
drug delivery applications [116]. In order to overcome mammalian cells.
the low solubility of CS in neutral pH, which is the Microcrystalline CS has been investigated as a gel
major drawback to use this type of polymer as a forming excipient [117]. Matrix granules of CS of
transfection agent, N-trimethylated and N-triethylated differing physicochemical properties loaded with
oligosaccharides have been synthesized [105]. either ibuprofen or paracetamol as model drugs have
Lee et al. [104] synthesized the hydrophobically been prepared. Varying the amount or molecular
modified CS containing 5.1 deoxycholic acid groups weight of the microcrystalline CS and to a lesser
per 100 anhydroglucose units by 1-ethyl-3-(3-dime- extent by the degree of deacetylation controlled
thylaminopropyl) carbodiimide (EDC)-mediated cou- release rate. Giunchedi et al. [59] prepared and
pling reaction as shown in Fig. 11. Since deoxycholic characterized a new derivative of CS: methyl
acid can form self-assemblies in aqueous media, it pyrrolidone CS. It randomly carries pyrrolidinone
was found that the modified CS also formed the self- groups covalently attached to the polysaccharide
aggregates. The self-aggregates were characterized by backbone. This CS derivative combines the biocom-

Fig. 11. A scheme of the coupling mechanism between chitosan and deoxycholic acid using 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide
(EDC) through amide linkage formation [taken from Ref. 104].
24 S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28

patibility of CS [118] and hydrophilic characteristics suggested that galactosylated CS microspheres could
of the pyrrolidinone moiety [119], being particularly be used for passive and active hepatic targeting.
susceptible to the hydrolytic action of lysozyme
[120]. The microparticles were characterized by
S.E.M., particle size analyzer, DSC and in vitro 7. Conclusions
ampicillin release. Drug release characteristics
depend upon the nature of CS used. Chitosan has the desired properties for safe use as a
Chen et al. [121] studied the modification of CS by pharmaceutical excipient. This has prompted accel-
coupling with linoleic acid (LA) through 1-ethyl-3-(3- erated research activities worldwide on chitosan micro
dimethylaminopropyl) carbodiimide-mediated reac- and nanoparticles as drug delivery vehicles. These
tion to increase its amphipathicity for improved systems have great utility in controlled release and
emulsification. The micelle formation of linoleic targeting studies of almost all class of bioactive
acid-modified CS in 0.1 M acetic acid solution was molecules as discussed in this review. Recently,
enhanced by O/W emulsification with methylene chitosan is also extensively explored in gene delivery.
chloride, an oil phase. Fluorescence spectra indicated However, studies toward optimization of process
that without emulsification, the self-aggregation of parameters and scale up from the laboratory to pilot
LA-CS occurred at the concentration of 1.0 g/L or plant and then, to production level are yet to be
above, and with emulsification, self-aggregation was undertaken. Majority of studies carried out so far are
greatly enhanced followed by a stable micelle only in in vitro conditions. More in vivo studies need
formation at 2.0 g/L. Addition of 1 M NaCl solution to be carried out. Chemical modifications of chitosan
promoted the self-aggregation of LA-CS particles are important to get the desired physicochemical
both with and without emulsification. The nanosize properties such as solubility, hydrophilicity, etc. The
micelles of LA-CS were formed ranging in size published literature indicates that in the near future,
between 200 and 600 nm. The LA-CS nanoparticles chitosan-based particulate systems will have more
were used to encapsulate the lipid soluble model commercial status in the market than in the past.
compound, retinal acetate, with 50% efficiency.
Chitosan was chemically modified [122] by graft
copolymerization of poly(ethylene glycol) diacrylate Acknowledgements
macromonomer onto CS backbone. Microspheres
based on chitosan and polymer grafted chitosan were Authors thank the University Grants Commission
prepared by a polymer dispersion technique. A (UGC), New Delhi, India for a major grant (F1-41/
comparative study in relation to structural deviation 2001/CPP-II) sanctioned to Karnatak University to
among CS and modified CS microspheres was establish Center of Excellence in Polymer Science.
evaluated. These chemically modified CS micropar-
ticles were hydrophilic in nature and formed aggre-
gates. Chitosan derivative with galactose groups was References
synthesized by introducing galactose group into the
amine group of CS [123]. The results indicated that [1] P.C. Berscht, B. Nies, A. Liebendorfer, J. Kreuter, Incorpo-
although acyl reaction on the part of amino groups of ration of basic fibroblast growth factor into methylpyrrolidi-
none chitosan fleeces and determination of the in vitro release
CS took place, the degree of galactosylated substitu-
characteristics, Biomaterials 15 (1994) 593 – 600.
tion was 20%. Crystallinity, solubility, stability and [2] L. Illum, Chitosan and its use as a pharmaceutical excipient,
other physical properties were different from CS. Pharm. Res. 15 (1998) 1326 – 1331.
Microspheres of CS and galactosylated CS were [3] V. Dodane, V.D. Vilivalam, Pharmaceutical applications of
prepared by the physical precipitation and coacerva- chitosan, Pharm. Sci. Technol. Today 1 (1998) 246 – 253.
tion techniques, respectively. Microspheres of CS and [4] O. Felt, P. Buri, R. Gurny, Chitosan: a unique polysaccharide
for drug delivery, Drug Dev. Ind. Pharm. 24 (1998) 979 – 993.
galactosylated CS were spherical in nature with an [5] K.D. Yao, T. Peng, Y.J. Yin, M.X. Xu, Microcapsules/
average diameter of 0.54 and 1.05 Am and an average microspheres related to chitosan, J. Macromol. Sci., Rev.
zeta potential of +17 and +15 mV, respectively. It was Macromol. Chem. Phys. C35 (1995) 155 – 180.
S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28 25

[6] H.S. Kas, Chitosan: properties, preparations and application [24] Y. Sawayanagi, N. Nambu, T. Nagai, Enhancement of
to microparticulate systems, J. Microencapsulation 14 (1997) dissolution properties of prednisolone from ground mix-
689 – 711. tures with chitin or chitosan, Chem. Pharm. Bull. 31
[7] R.A.A. Muzzarelli, C. Jeuniauk, G.W. Gooday, Chitin in (1983) 2507 – 2509.
Nature and Technology, Plenum, New York, 1986. [25] J.M. Gallo, E.E. Hassan, Receptor-mediated magnetic
[8] G. Sjak-Braek, T. Anthonsen, P. Sandford, Chitin and carriers: basis for targeting, Pharm. Res. 5 (1988) 300 – 304.
Chitosan, Elsevier, New York, 1992. [26] E.E. Hassan, R.C. Parish, J.M. Gallo, Optimized formulation
[9] T. Sannan, K. Kurita, Y. Iwakura, Studies on chitin, 2. Effect of magnetic chitosan microspheres containing the anticancer
of deacetylation on solubility, Makromol. Chem. 177 (1976) agent, oxantrazole, Pharm. Res. 9 (1992) 390 – 397.
3589 – 3600. [27] P. Artursson, T. Lindmark, S.S. Davis, L. lllum, Effect
[10] S. Nicol, Life after death for empty shells, New Sci. 129 of chitosan on the permeability of monolayers of
(1991) 46 – 48. intestinal epithelial cells (CACO-2), Pharm. Res. 11
[11] K. Arai, T. Kinumaki, T. Fujita, Toxicity of chitosan, Bull. (1994) 1358 – 1361.
Tokai Reg. Fish Lab. 43 (1968) 89 – 94. [28] J. Akbuga, G. Durmaz, Preparation and evaluation of cross-
[12] T. Chandy, C.P. Sharma, Chitosan as a biomaterial, Biomater. linked chitosan microspheres containing furosemide, Int. J.
Artif. Org. 18 (1990) 1 – 24. Pharm. 111 (1994) 217 – 222.
[13] W.M. Hou, S. Miyazaki, M. Takada, T. Komai, Pharmaceut- [29] B.C. Thanoo, M.C. Sunny, A. Jayakrishnan, Cross-linked
ical application of biomedical polymers. Part XVI. Sustained chitosan microspheres: preparation and evaluation as a matrix
release of indomethacin from chitosan, Chem. Pharm. Bull. for the controlled release of pharmaceuticals, J. Pharm.
33 (1985) 3986 – 3992. Pharmacol. 44 (1992) 283 – 286.
[14] S. Miyazaki, K. Ishii, T. Nadai, Pharmaceutical application of [30] K.S. Soppimath, A.R. Kulkarni, T.M. Aminabhavi, Con-
biomedical polymers. Part IV. The use of chitin and chitosan trolled release of antihypertensive drug from the interpene-
as drug carriers, Chem. Pharm. Bull. 29 (1981) 3067 – 3069. trating network poly(vinyl alcohol) guar gum hydrogel
[15] T. Handa, A. Kasai, H. Takenaka, S.Y. Lin, Y. Ando, Novel microspheres, J. Biomater. Sci., Polym. Ed. 11 (2000) 27 – 43.
method for the preparation of controlled-release theophylline [31] K.S. Soppimath, A.R. Kulkarni, T.M. Aminabhavi, Water
granules coated with a polyelectrolyte complex of sodium transport and drug release study of cross-linked guar gum
polyphosphate-chitosan, J. Pharm. Sci. 74 (1985) 264 – 268. grafted polyacrylamide hydrogel microspheres for the con-
[16] S. Miyazaki, H. Yamaguchi, C. Yokouchi, M. Takada, W.-M. trolled release application, Eur. J. Pharm. Biopharm. 53
Hou, Sustained-release and intragastric-floating granules of (2002) 87 – 98.
indomethacin using chitosan in rabbits, Chem. Pharm. Bull. [32] A.R. Kulkarni, K.S. Soppimath, T.M. Aminabhavi, Con-
36 (1988) 4033 – 4038. trolled release of cefadroxil using sodium alginate inter-
[17] Y. Sawayanagi, N. Nambu, T. Nagai, Use of chitosan for penetrating network with gelatin/egg albumin, Eur. J. Pharm.
sustained-release preparations of water-soluble drugs, Chem. Biopharm. 51 (2001) 127 – 133.
Pharm. Bull. 30 (1982) 4213 – 4215. [33] S.G. Kumbar, A.R. Kulkarni, T.M. Aminabhavi, Cross-
[18] S. Shiraishi, T. Imai, M. Otagiri, Controlled release of linked chitosan microspheres for encapsulation of diclofenac
indomethacin by chitosan– polyelectrolyte complex: optimi- sodium: effect of cross-linking agent, J. Microencapsulation
zation and in vivo/in vitro evaluation, J. Control. Release 25 19 (2002) 173 – 180.
(1993) 217 – 225. [34] S.G. Kumbar, T.M. Aminabhavi, Synthesis and character-
[19] C.M. Lehr, J.A. Bouwstra, E.H. Schacht, H.E. Junginger, In ization of modified chitosan microspheres: effect of the
vitro evaluation of mucoadhesive properties of chitosan and grafting ratio on the controlled release of nifedipine through
some other natural polymers, Int. J. Pharm. 78 (1992) 43 – 48. microspheres, J. Appl. Polym. Sci. 89 (2003) 2940 – 2949.
[20] H.L. Luehen, C.M. Lehr, C.O. Rentel, A.B.J. Noach, A.G. [35] K.S. Soppimath, T.M. Aminabhavi, A.R. Kulkarni, W.E.
Boer, J.C. Verhoef, H.E. Junginger, Bioadhesive polymers for Rudzinski, Biodegradable polymeric nanoparticles as drug
the peroral delivery of peptide drugs, J. Control. Release 29 delivery devices, J. Control. Release 70 (2001) 1 – 20.
(1994) 329 – 338. [36] D.K. Singh, A.R. Ray, Biomedical applications of chitin,
[21] L. Illum, N.F. Farraj, S.S. Davis, Chitosan as a novel nasal chitosan and their derivatives, J. Macromol. Sci., Rev.
delivery system for peptide drugs, Pharm. Res. 11 (1994) Macromol. Chem. Phys. C40 (2000) 69 – 83.
1186 – 1189. [37] A.K. Singla, M. Chawla, Chitosan: some pharmaceutical and
[22] T. Imai, S. Shiraishi, H. Saito, M. Otagiri, Interaction of biological aspects an update, J. Pharm. Pharmacol. 53 (2001)
indomethacin with low molecular weight chitosan, and 1047 – 1067.
improvements of some pharmaceutical properties of indome- [38] W.G. Liu, K.D. Yao, Chitosan and its derivatives– a promis-
thacin by low molecular weight chitosans, Int. J. Pharm. 67 ing non-viral vector for gene transfection, J. Control. Release
(1991) 11 – 20. 83 (2002) 1 – 11.
[23] I. Genta, F. Pavanetto, B. Conti, P. Giunchedi, U. Conte, [39] R. Hejazi, M. Amiji, Chitosan-based gastrointestinal delivery
Spray-drying for the preparation of chitosan microspheres, systems, J. Control. Release 89 (2003) 151 – 165.
Proc. Int. Symp. Control. Release Bioact. Mater. 21 (1994) [40] V.R. Sinha, A.K. Singla, S. Wadhawan, R. Kaushik, R.
616 – 617. Kumria, K. Bansal, S. Dhawan, Chitosan microspheres as
26 S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28

a potential carrier for drugs, Int. J. Pharm. 274 (2004) oral-administration controlled release of metoclopramide,
1 – 33. Eur. J. Pharm. Biopharm. 48 (1999) 149 – 155.
[41] J. Kreuter, Nanoparticles, in: J. Kreuter (Ed.), Colloidal Drug [57] X.Y. Shi, T.W. Tan, Preparation of chitosan/ethylcellulose
Delivery Systems, Marcel Dekker, New York, 1994, pp. complex microcapsule and its application in controlled
219 – 342. release of vitamin D-2, Biomaterials 23 (2002) 4469 – 4473.
[42] L. Brannon Peppas, Recent advances on the use of [58] C.W. Bishop, J.C. Knutson, C.R. Valliere, Method of treating
biodegradable microparticles and nanoparticles in the con- prostatic disease using delayed and/or sustained release
trolled drug delivery, Int. J. Pharm. 116 (1995) 1 – 9. vitamin D formulation, US Pat. 5 795 882, 1998.
[43] P. Couvreur, L. Grislain, V. Lenaerts, F. Brasseur, P. Guiot, [59] P. Giunchedi, I. Genta, B. Conti, R.A.A. Muzzarelli, U.
in: P. Guiot, P. Couvreur (Eds.), Polymeric Nanoparticles and Conte, Preparation and characterization of ampicillin loaded
Microspheres, CRC Press, Boca Raton, FL, 1986. methylpyrrolidinone chitosan and chitosan microspheres,
[44] J. Akbuga, G. Durmaz, Preparation and evaluation of cross- Biomaterials 19 (1998) 157 – 161.
linked chitosan microspheres containing furosemide, Int. J. [60] M.L. Lorenzo-Lamosa, C. Remunan-Lopez, J.L. Vila-Jato,
Pharm. 11 (1994) 217 – 222. M.J. Alonso, Design of microencapsulated chitosan micro-
[45] A.A. Al-Helw, A.A. Al-Angary, G.M. Mahrous, M.M. Al- spheres for colonic drug delivery, J. Control. Release 52
Dardari, Preparation and evaluation of sustained release (1998) 109 – 118.
cross-linked chitosan microspheres containing phenobarbi- [61] Y.C. Huang, M.K. Yeh, C.H. Chiang, Formulation factors in
tone, J. Microencapsulation 15 (1998) 373 – 382. preparing BTM-chitosan microspheres by spray drying
[46] E.B. Denkbas, M. Seyyal, E. Piskin, 5-Fluorouracil loaded method, Int. J. Pharm. 242 (2002) 239 – 242.
chitosan microspheres for chemoembolization, J. Micro- [62] H. Tokumitsu, H. Ichikawa, Y. Fukumori, Chitosan– gado-
encapsulation 16 (1998) 741 – 749. pentetic acid complex nanoparticles for gadolinium neutron
[47] C. Sankar, M. Rani, A.K. Srivastava, B. Mishra, Chitosan capture therapy of cancer: preparation by novel emulsion-
based pentazocine microspheres for intranasal systemic droplet coalescence technique and characterization, Pharm.
delivery: development and biopharmaceutical evaluation, Res. 16 (1999) 1830 – 1835.
Pharmazie 56 (2001) 223 – 226. [63] A. Polk, B. Amsden, K.D. Yao, T. Peng, M.F.A. Goosen,
[48] S.R. Jameela, T.V. Kumary, A.V. Lal, A. Jayakrishnan, Controlled release of albumin from chitosan-alginate micro-
Progesterone-loaded chitosan microspheres: a long acting capsules, J. Pharm. Sci. 83 (1994) 178 – 185.
biodegradable controlled delivery system, J. Control. Release [64] L.S. Liu, S.Q. Liu, S.Y. Ng, M. Froix, T. Ohno, J. Heller,
52 (1998) 17 – 24. Controlled release of interleukin-2 for tumor immunotherapy
[49] F. Bugamelli, M.A. Raggi, I. Orienti, V. Zecchi, Controlled using alginate:chitosan porous microspheres, J. Control.
insulin release from chitosan microparticles, Arch. Pharm. Release 43 (1997) 65 – 74.
331 (1998) 133 – 138. [65] Y. Kawashima, T. Handa, H. Takenaka, S.Y. Lin, Y. Ando,
[50] K. Nishimura, S. Nishimura, H. Seo, N. Nishi, S. Tokura, I. Novel method for the preparation of controlled-release
Azuma, Macrophage activation with multiporous beads theophylline granules coated with a polyelectrolyte complex
prepared from partially deacetylated chitin, J. Biomed. Mater. of sodium polyphosphate-chitosan, J. Pharm. Sci. 74 (1985)
Res. 20 (1986) 1359 – 1372. 264 – 268.
[51] A. Berthod, J. Kreuter, Chitosan microspheres– improved [66] Y. Kawashima, T. Handa, A. Kasai, H. Takenaka, S.Y. Lin,
acid stability and change in physicochemical properties by The effect of thickness and hardness of the coating film on
cross-linking, Proc. Int. Symp. Control. Release Bioact. the drug release of theophylline granules, Chem. Pharm.
Mater. 23 (1996) 369 – 370. Bull. 33 (1985) 2469 – 2474.
[52] S. Ozbas-Turan, J. Akbuga, C. Aral, Controlled release of [67] R. Bodmeier, K.H. Oh, Y. Pramar, Preparation and evaluation
interleukin-2 from chitosan microspheres, J. Pharm. Sci. 91 of drug-containing chitosan beads, Drug Dev. Ind. Pharm. 15
(2002) 124 – 125. (1989) 1475 – 1494.
[53] H.Q. Mao, K. Roy, V.L. Troung-Le, K.A. Janes, K.Y. Lim, Y. [68] S. Shirashi, T. Imai, M. Otagiri, Controlled release of
Wang, J.T. August, K.W. Leong, Chitosan DNA nano- indomethacin by chitosan– polyelectrolyte complex: optimi-
particles as gene delivery carriers: synthesis, characterization zation and in vivo: in vitro evaluation, J. Control. Release 25
and transfection efficiency, J. Control. Release 70 (2001) (1993) 217 – 225.
399 – 421. [69] A.D. Sezer, J. Akbuga, Controlled release of piroxicam from
[54] P. He, S.S. Davis, L. Illum, Chitosan microspheres prepared chitosan beads, Int. J. Pharm. 121 (1995) 113 – 116.
by spray drying, Int. J. Pharm. 187 (1999) 53 – 65. [70] Z. Aydin, J. Akbuga, Chitosan beads for the delivery of
[55] B. Conti, T. Modena, I. Genta, P. Perugini, C. Decarro, F. salmon calcitonin: preparation and characteristics, Int. J.
Pavanetto, Microencapsulation of cetylpyridinium chloride Pharm. 131 (1996) 101 – 103.
with a bioadhesive polymer, Proc. Int. Symp. Control. [71] P. Calvo, C. Remunan-Lopez, J.L. Vila-Jata, M.J. Alonso,
Release Bioact. Mater. 25 (1998) 822 – 823. Chitosan and chitosan: ethylene oxide-propylene oxide block
[56] A. Ganza-Gonzalez, S. Anguiano-Igea, F.J. Otero-Espinar, copolymer nanoparticles as novel carriers for protein and
J.B. Mendez, Chitosan and chondroitin microspheres for vaccines, Pharm. Res. 14 (1997) 1431 – 1436.
S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28 27

[72] P. Calvo, C. Remunan-Lopez, J.L. Vila-Jata, M.J. Alonso, [90] P.L. Ritger, N.A. Peppas, A simple equation for description
Novel hydrophilic chitosan-polyethylene oxide nanopar- of solute release. II. Fickian and anomalous release from
ticles as protein carriers, J. Appl. Polym. Sci. 63 (1997) swellable devices, J. Control. Release 5 (1987) 37 – 42.
125 – 132. [91] N.A. Peppas, R.W. Korsmeyer, in: N.A. Peppas (Ed.),
[73] X.Z. Shu, K.J. Zhu, A novel approach to prepare tripoly- Hydrogels in Medicine and Pharmacy, vol. 3, CRC Press,
phosphate/chitosan complex beads for controlled release drug Boca Raton, FL, 1987, p. 103.
delivery, Int. J. Pharm. 201 (2000) 51 – 58. [92] I. Orienti, K. Aiedeh, E. Gianasi, V. Bertasi, V. Zecchi,
[74] R. Fernandez-Urrusuno, P. Cavlo, C. Remunan-Lopez, J.L. Indomethacin loaded chitosan microspheres. Correlation
Vila-Jato, M.J. Alonso, Enhancement of nasal absorption of between the erosion process and release kinetics, J. Micro-
insulin using chitosan nanoparticles, Pharm. Res. 16 (1999) encapsulation 13 (1996) 463 – 472.
1576 – 1581. [93] D. Pantaleone, M. Yalpani, M. Scollar, Advances in chitin
[75] Y. Pan, Y. Li, H. Zhao, J. Zheng, H. Xu, G. Wei, J. Hao, F. and chitosan, in: C.J. Brine, P.A. Sandford, J.P. Zizakis,
Cui, Chitosan nanoparticles improve the intestinal absorption Proceed. 5th Int. Conf. Chitin and Chitosan, Princeton, New
of insulin in vivo, Int. J. Pharm. 249 (2002) 139 – 147. York, 1991.
[76] Y. Xu, Y. Du, Effect of molecular structure of chitosan on [94] H. Zhang, I.A. Alsarra, S.H. Neau, An in vitro evaluation of a
protein delivery properties of chitosan nanoparticles, Int. J. chitosan-containing multiparticulate system for macromole-
Pharm. 250 (2003) 215 – 226. cule delivery to the colon, Int. J. Pharm. 239 (2002) 197 – 205.
[77] J.A. Ko, H.J. Park, S.J. Hwang, J.B. Park, J.S. Lee, [95] I. Genta, M. Costantini, A. Asti, B. Conti, L. Montanari,
Preparation and characterization of chitosan microparticles Influence of glutaraldehyde on drug release and mucoadhe-
intended for controlled drug delivery, Int. J. Pharm. 249 sive properties of chitosan microspheres, Carbohydr. Polym.
(2002) 165 – 174. 36 (1998) 81 – 88.
[78] Y.S. Leong, F. Candau, Inverse microemulsion polymer- [96] I.M. van der Lubben, F.A.J. Konings, G. Borchard, J.C.
ization, J. Phys. Chem. 86 (1982) 2269 – 2271. Verhoef, H.E. Junginger, In vivo uptake of chitosan micro-
[79] A. Maitra, Determination of size parameters of water– particles by murine Peyer’s patches: visualization studies
Aerosol OT– oil reverse micelles from their nuclear magnetic using confocal laser scanning microscopy and immuno-
resonance data, J. Phys. Chem. 88 (1984) 5122 – 5125. histochemistry, J. Drug Target. 9 (2001) 39 – 47.
[80] P.L. Luisi, M. Giomini, M.P. Pileni, B.H. Robinson, Reverse [97] I.M. van der Lubben, G. Kersten, M.M. Fretz, C. Beuvery,
micelles as hosts for proteins and small molecules, Biochim. J.C. Verhoef, H.E. Junginger, Chitosan microparticles for
Biophys. Acta 947 (1988) 209 – 246. mucosal vaccination against diphtheria: oral and nasal
[81] S. Mitra, U. Gaur, P.C. Ghosh, A.N. Maitra, Tumor targeted efficacy studies in mice, Vaccine 21 (2003) 1400 – 1408.
delivery of encapsulated dextran– doxorubicin conjugate [98] I.M. van der Lubben, J.C. Verhoef, A.C. van Aelst, G.
using chitosan nanoparticles as carrier, J. Control. Release Borchard, H.E. Junginger, Chitosan microparticles for oral
74 (2001) 317 – 323. vaccination: preparation, characterization and preliminary in
[82] S.A. Agnihotri, T.M. Aminabhavi, Controlled release of vivo uptake studies in murine Peyer’s patches, Biomaterials
clozapine through chitosan microparticles prepared by a 22 (2001) 687 – 694.
novel method, J. Control. Release 96 (2004) 245 – 259. [99] F. Shikata, H. Tokumitsu, H. Ichikawa, Y. Fukumori, In vitro
[83] J. Akbuga, N. Bergisadi, Effect of formulation variables on cellular accumulation of gadolinium incorporated into chito-
cis-platin loaded chitosan microsphere properties, J. Micro- san nanoparticles designed for neutron-capture therapy of
encapsulation 16 (1999) 697 – 703. cancer, Eur. J. Pharm. Biopharm. 53 (2002) 57 – 63.
[84] S.R. Jameela, A. Misra, A. Jayakrishnan, Cross-linked [100] H. Tokumitsu, J. Hiratsuka, Y. Sakurai, T. Kobayashi, H.
chitosan microspheres as carriers for prolonged delivery of Ichikawa, Y. Fukumori, Gadolinium neutron-capture therapy
macromolecular drugs, J. Biomater. Sci., Polym. Ed. 6 (1994) using novel gadopentetic acid-chitosan complex nanopar-
621 – 632. ticles: in vivo growth suppression of experimental melanoma
[85] R. Hejazi, M. Amiji, Stomach-specific anti-H. pylori therapy. solid tumor, Cancer Lett. 150 (2000) 177 – 182.
I: preparation and characterization of tetracyline-loaded [101] S.R. Jameela, P.G. Latha, A. Subramoniam, A. Jayakrishnan,
chitosan microspheres, Int. J. Pharm. 235 (2002) 87 – 94. Antitumor activity of mitoxantrone-loaded chitosan micro-
[86] D.K. Kweon, D.W. Kang, Drug-release behavior of chitosan- spheres against Ehrlich ascites carcinoma, J. Pharm. Phar-
g-poly(vinyl alcohol) copolymer matrix, J. Appl. Polym. Sci. macol. 48 (1996) 685 – 688.
74 (1999) 458 – 464. [102] S.R. Jameela, A. Jayakrishnan, Glutaraldehyde cross-linked
[87] T. Higuchi, Mechanism of sustained action medication, J. chitosan microspheres as a long-acting biodegradable drug-
Pharm. Sci. 52 (1963) 1145 – 1149. delivery vehicle-studies on the in-vitro release of mitoxan-
[88] C. Washington, Drug release from microdisperse systems: a trone and in-vivo degradation of microspheres in rat muscle,
critical review, Int. J. Pharm. 58 (1990) 1 – 12. Biomaterials 16 (1995) 769 – 775.
[89] R.H. Guy, J. Hadgraft, I.W. Kellaway, M.J. Taylor, Calcu- [103] K.A. Janes, M.P. Fresneau, A. Marazuela, A. Fabra, M.J.
lations of drug release rates from spherical particles, Int. J. Alonso, Chitosan nanoparticles as delivery systems for
Pharm. 11 (1982) 199 – 207. doxorubicin, J. Control. Release 73 (2001) 255 – 267.
28 S.A. Agnihotri et al. / Journal of Controlled Release 100 (2004) 5–28

[104] K.Y. Lee, I.C. Kwon, Y.H. Kim, W.H. Jo, S.Y. Jeong, [114] S.H. Chiou, W.T. Wu, Y.Y. Huang, T.W. Chung, Effects of
Preparation of chitosan self-aggregates as a gene delivery the characteristics of chitosan on controlling drug release of
system, J. Control. Release 51 (1998) 213 – 220. chitosan coated PLLA microspheres, J. Microencapsulation
[105] B.I. Florea, M. Thanou, M. Geldof, C. Meaney, H.E. 18 (2001) 613 – 625.
Junginger, G. Borchard, Modified chitosan oligosaccharides [115] M. Thanou, J.C. Verhoef, P. Marbach, H.E. Junginger,
as transfection agents for gene therapy of cystic fibrosis, Intestinal absorption of octreotide: N-trimethyl chitosan
Proc. Int. Symp. Control. Release Bioact. Mater. 27 (2000) chloride (TMC) ameliorates the permeability and absorption
846 – 847. properties of the somatostatin analogue in vitro and in vivo, J.
[106] P. Erbacher, S. Zou, T. Beuinger, A.M. Steffan, J.S. Remy, Pharm. Sci. 89 (2000) 951 – 957.
Chitosan-based vector/DNA complexes for gene delivery: [116] K. Aiedeh, M.O. Taha, Synthesis of chitosan succinate and
biophysical characteristics and transfection ability, Pharm. chitosan phthalate and their evaluation as suggested matrices
Res. 15 (1998) 1332 – 1339. in orally administered, colon-specific drug delivery systems,
[107] Y.K. Park, Y.H. Park, B.A. Shin, E.S. Choi, Y.R. Park, T. Archiv.der Pharmazie 332 (1999) 103 – 107.
Akaike, C.S. Cho, Galactosylated chitosan graft-dextran as [117] M. Sakkinen, U. Seppala, P. Heinanen, M. Marvola, In vitro
hepatocyte-targeting DNA carrier, J. Control. Release 69 evaluation of microcrystalline chitosan (MCCh) as gel-
(2000) 97 – 108. forming excipient in matrix granules, Eur. J. Pharm.
[108] J.L. Chew, C.B. Wolfowicz, H.Q. Mao, K.W. Leong, K.Y. Biopharm. 54 (2002) 33 – 40.
Chua, Chitosan nanoparticles containing plasmid DNA [118] R.A.A. Muzzarelli, V. Baldassarre, F. Conti, G. Gazzanelli, V.
encoding house dust mite allergen, Der p 1 for oral Vasi, P. Ferrara, G. Biagini, The biological activity of chitosan:
vaccination in mice, Vaccine 21 (2003) 2720 – 2729. ultrastructural study, Biomaterials 8 (1988) 247 – 252.
[109] Z. Cui, R.J. Mumper, Chitosan-based nanoparticles for [119] R.A.A. Muzzarelli, Depolymerization of methylpyrrolidinone
topical genetic immunization, J. Control. Release 75 (2001) chitosan by lysozyme, Carbohydr. Polym. 19 (1992) 29 – 34.
409 – 419. [120] R.A.A. Muzzarelli, In vivo biochemical significance of
[110] G. Borchard, Chitosans for gene delivery, Adv. Drug Deliv. chitin-based medical items, in: S. Dumitriu, M. Szycher
Rev. 52 (2001) 145 – 150. (Eds.), Polymeric Materials for Biomedical Applications,
[111] B. Conti, P. Giunchedi, I. Genta, U. Conte, The preparation Marcel Dekker, New York, 1992, pp. 179 – 198.
and in vivo evaluation of the wound-healing properties of [121] X.G. Chen, C.M. Lee, H.J. Park, O/W emulsification for the
chitosan microspheres, STP Pharma. Sci. 10 (2000) 101 – 104. self-aggregation and nanoparticle formation of linoleic acids
[112] A.M. De Campos, A. Sanchez, M.J. Alonso, Chitosan modified chitosan in the aqueous system, J. Agric. Food
nanoparticles: a new vehicle for the improvement of the Chem. 51 (2003) 3135 – 3139.
delivery of drugs to the ocular surface. Application to [122] K.L. Shanta, D.R.K. Harding, Synthesis and characterization
cyclosporin A, Int. J. Pharm. 224 (2001) 159 – 168. of chemically modified chitosan microspheres, Carbohydr.
[113] T. Chandy, R.F. Wilson, G.H.R. Rao, G.S. Das, Changes in Polym. 48 (2002) 247 – 253.
cisplatin delivery due to surface-coated poly(lactic acid)- [123] C. Zhang, Q. Ping, Y. Ding, Y. Cheng, J. Shen, Synthesis,
poly(q-caprolactone) microspheres, J. Biomater. Appl. 16 characterization and microsphere formation of galactosylated
(2002) 275 – 291. chitosan, J. Appl. Polym. Sci. 91 (2004) 659 – 665.

Potrebbero piacerti anche