You are on page 1of 16

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/327021895

Chitosan-based nanoparticles as drug delivery systems: A review on two


decades of research

Article  in  Journal of Drug Targeting · August 2018


DOI: 10.1080/1061186X.2018.1512112

CITATIONS READS
101 786

3 authors:

Sweet Naskar Ketousetuo Kuotsu


Jadavpur University Jadavpur University
10 PUBLICATIONS   230 CITATIONS    33 PUBLICATIONS   1,548 CITATIONS   

SEE PROFILE SEE PROFILE

Suraj Sharma
Jadavpur University
7 PUBLICATIONS   223 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Toxicity of glutaraldehyde in the preparation of nanoparticles. View project

All content following this page was uploaded by Sweet Naskar on 22 May 2021.

The user has requested enhancement of the downloaded file.


JOURNAL OF DRUG TARGETING
2019, VOL. 27, NO. 4, 379–393
https://doi.org/10.1080/1061186X.2018.1512112

REVIEW ARTICLE

Chitosan-based nanoparticles as drug delivery systems: a review on two decades


of research
Sweet Naskar, Ketousetuo Kuotsu and Suraj Sharma
Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India

ABSTRACT ARTICLE HISTORY


Chitosan (CS) is one of the most functional natural biopolymer widely used in the pharmaceutical field due Received 8 May 2018
to its biocompatibility and biodegradability. These privileges lead to its application in the synthesis of nano- Revised 7 August 2018
particles for the drug during the last two decades. This article gives rise to a general review of the different Accepted 8 August 2018
chitosan nanoparticles (CSNPs) preparation techniques: Ionic gelation, emulsion cross-linking, spray-drying,
KEYWORDS
emulsion-droplet coalescence method, nanoprecipitation, reverse micellar method, desolvation method, Chitosan nanoparticles;
modified ionic gelation with radial polymerisation and emulsion solvent diffusion, from the point of view of drug delivery;
the methodological and mechanistic aspects involved. The physicochemical behaviour of CSNPs including Pharmaceutical applications;
drug loading, drug release, particles size, zeta potential and stability are briefly discussed. This review also Derivatives of Chitosan
directs to bring an outline of the major applications of CSNPs in drug delivery according to drug and route nanoparticles; Chitosan
of administration. Finally, derivatives of CSNPs and CS nano-complexes are also discussed. nanocomplexes

Abbreviations: CSF: Cerebrospinal fluid; 5-FU: 5- Fluorouracil; CyA: Cyclosporin A; FITC-BSA: Fluorescein
isothiocyanate labeled bovine serum albumin; pDNA: Plasmid deoxyribonucleic acid; CS- NAC: Chitosan N-
acetyl-L-cysteine; Glycol CS-TGA: Glycol chitosan thioglycolic acid; CS-TGA: Chitosan thioglycolic acid; CS-
TBA: Chitosan $- thiobutylamidine; pSEAP: Recombinant secreted alkaline phosphate; OVA: Ovalbumin;
Thiolated HA: Thiolated hyaluronic acid; SVN: Survivin; SiRNA: Small interfering ribonucleic acid.

Introduction crystalline structure due to the inter and intra-molecular hydrogen


bonds between the hydroxyl and amino groups. Due to the pres-
Biodegradable polymeric nanoparticles (NPs) can act as efficient
ence of N-acetyl groups, CS shows a slight degree of hydrophobic
drug delivery vehicles for controlled and targeted drug [1].
behaviour [12–18].
Chitosan (CS) is a polysaccharide, used as NPs in the pharmaceut-
ical field since two decades ago [2]. CS is a biodegradable,
biocompatible and non-toxic biopolymer, made up of b-1 ! 4 Preparation of chitosan nanoparticles (CSNPs)
linked 2-amino-2-deoxy-glucopyranose and 2-acetamido-2-deoxy-
b-D-glucopyranose residues. Chitin, a biopolymer extracted from CSNPs have been fully recorded in the literature as a carrier sys-
the exoskeleton of crustaceans, such as crabs and shrimp is used tem for various drug delivery systems.Since first reported in 1994
to prepare CS, is produced by alkaline N-deacetylation [3–5]. CS by Ohya et al. [19], various techniques have been used to
shows its antibacterial activity only in an acidic medium above pH develop CSNPs.
6.5 [6].
Ionic gelation
Source and physicochemical properties of CS In this method, firstly, CS is dissolved in an aqueous acidic solu-
CS is a polysaccharide composed of b-1, 4-linked D-glucosamine tion to obtain the cation of CS. After that, this solution is added
residue. It is derived from the deacetylation of chitin, a naturally dropwise to anionic tripolyphosphate (TPP) solution under con-
occurring polymer found in the exoskeletons of crustaceans and stant stirring. CS undergoes ionic gelation due to the complex-
insects [6]. CS has been officially agreed by FDA for the use of ation between polyanion TPP and cationic CS by electrostatic
wound dressings as well as in dietary applications in Japan, Italy forces and precipitates to form spherical particles [20–36]. The
and Finland [7]. It is produced on a commercial scale by exhaust- method is schematically represented in Figure 1.
ive deacetylation of chitin with the concentrated solution of
sodium hydroxide [8,9]. CS, a non-antigenic hydrophilic polymer,
Emulsion cross-linking
containing one amino group and two hydroxyl groups in the
restating hexosaminic residue [10,11]. The amino groups make CS In this method, firstly, an aqueous CS solution is emulsified in the
a natural polyelectrolyte that readily dissolves in an acidic solu- oil phase to prepare water-in-oil (W/O) emulsion and after that
tion. The insolubility of CS in the solution and the degree of aqueous droplets are stabilised using a suitable surfactant. Then,
acetylation (DA) effect on the density of it. CS possesses a the stable emulsion is cross-linked with the most versatile cross-

CONTACT Ketousetuo Kuotsu ketousetuoju@yahoo.in Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
This article has been republished with minor changes. These changes do not impact the academic content of the article.
ß 2018 Informa UK Limited, trading as Taylor & Francis Group
380 S. NASKAR ET AL.

Figure 1. Representation of preparation of CSNPs by ionic gelation method.

Figure 4. Representation of preparation of CSNPs by emulsion-droplet coales-


cence method.
Figure 2. Representation of preparation of CSNPs by emulsion crosslink-
ing method. Spray-drying
In this method, CSNPs are prepared by use of a nanospray dryer.
The method depends on the drying of atomised droplets in a hot
air stream. Firstly, a CS solution is prepared by dissolving purified
CS powder in an acetic acid solution and stores it until overnight.
Small droplets are formed due to atomisation. The solvent is
evaporated from small droplets instantaneously leading to the
formation of NPs. This method is schematically shown in
Figure 3 [39–42].

Emulsion droplet coalescence method


In this method, at first, two stable emulsions are formed. At firstly,
a stable emulsion containing an aqueous solution of CS along
with drug is brought about in liquid paraffin oil. After that,
another stable emulsion containing CS aqueous solution of NaOH
is carried out in liquid paraffin oil. Then two emulsions are mixed
under high-speed stirring. When these two emulsions are mixed,
then each emulsion droplets would come into conflict at random
and coalesce, as a result of that precipitating CS droplet to form
small size particles [43,44]. The method is schematically shown in
Figure 4.

Nanoprecipitation
Figure 3. Representation of preparation of CSNPs by spray drying method. In this method, firstly, CS is dissolved in a suitable solvent to form
the diffusing phase. This phase is then added to the dispersing
linking agent, i.e. glutaraldehyde. Here, cross-linking reaction took phase i.e. methanol under magnetic stirring condition. The diffus-
place between the amino groups of CS with the aldehyde groups ing phase is added to the dispersing phase by means of a needle
of glutaraldehyde and precipitates to form particles [19,37,38]. positioned 2 cm above the surface at 0.86 ml min1 using a peri-
This method is schematically shown in Figure 2. staltic pump. Then the addition of a very small amount of
JOURNAL OF DRUG TARGETING 381

Figure 5. Representation of preparation of CSNPs by nanoprecipitation method.

Figure 7. Representation of preparation of CSNPs by desolvation method.

Figure 6. Representation of preparation of CSNPs by reverse micellar method.


Figure 8. Representation of preparation of CSNPs by modified ionic gelation with
radical polymerisation method.
polysorbate-80 to the non-solvent phase provides smaller NPs
regardless of the non-solvent to solvent volume ratio [45]. The
method is schematically represented in Figure 5. added to the formulation, in order to harden the NPs [51]. This
method is schematically shown in Figure 7.

Reverse micellisation method


Modified ionic gelation with radical polymerisation
In reverse micellisation method, W/O droplets are known as
In this method, under stirring condition, an aqueous acrylic acid
reverse micelles [46,47]. Firstly, a W/O emulsion is prepared using
monomer solution is added to an aqueous CS solution at room
a lipophilic surfactant [sodium bis(ethyl hexyl) sulfosuccinate or
temperature. Sometimes, polyethylene glycol (PEG) or polyether
cetyl trimethyl ammonium bromide] that is dissolved in an appro-
(PEG–polypropylene glycol–PEG) is also added to the reaction
priate organic solvent, such as n-hexane. After that, an aqueous
medium, either separately into the monomer solution or the fol-
CS solution, drug and glutaraldehyde are added to the organic
lowing mixing with CS. Ionic gelation occurs due to ionic inter-
phase under continuous stirring to avoid turbidity. Finally, extrac-
action between the cation of CS and anion of acrylic acid
tion of NPs is done [48–50]. This method is schematically shown
monomer and radical polymerisation of the latter is started with
in Figure 6. potassium persulfate under stream nitrogen at 60–70  C. After, the
polymerisation reaction the formed NPs suspension is allowed to
settle overnight. Finally, the dialysis process is used to remove
Desolvation/simple coacervation/phase separation
unreacted monomers [58–60]. This method is schematically shown
In this method, at first, a CS solution is prepared using a suitable in Figure 8.
solvent. After that, the solvent competing agent of greater hydro-
philicity (e.g. sodium sulphate) is added to this CS solution. When
saltly enters in contact with the aqueous environment of CS solu- Emulsion solvent diffusion
tion then a progressive elimination of salvation water surrounding In this method, firstly, the organic phase (e.g. methylene chloride
CS occurs as a result of the higher affinity of water for the salt. and acetone) containing the hydrophobic drug is added to an
This step leads to the polymer insolubilisation and as a result, pre- aqueous solution containing CS and a stabiliser (e.g. poloxamer
cipitation occurs [51–53]. Sodium sulphate [19,54–56] and acetone and lecithin) to prepare an oil-in-water (O/W) emulsion under stir-
[57] are used as precipitating agents in this process. Polysorbate- ring condition. Then the formed O/W emulsion is subjected to
80 is used as a stabilising agent in the preparation medium, to high-pressure homogenisation and methylene chloride is removed
stabilise the nanoparticle suspension. After that, glutaraldehyde is under reduced pressure at room temperature. At this stage,
382 S. NASKAR ET AL.

NPs by ionic gelation method. The antibacterial activity of the pre-


pared NPs was evaluated against Escherichia coli MTCC 443
by them.

Citric acid
Thermal cross-linking is another aspect of physical cross-linking in
which citric acid (CA), a commonly used cross-linking agent is
added to an aqueous CS acidic solution in a constant molar ratio
between CA and CS [77]. Bagheri et al. [78] modified CS with carb-
oxyl functional groups using CA as a cross-linking agent at the dif-
Figure 9. Representation of preparation of CSNPs by emulsion solvent diffu- ferent molar ratio of CS to CA.
sion method.

acetone is diffused into the aqueous phase, decreasing CS solubil- Other crosslinkers
ity and thus, NPs are formed by polymer precipitation. An extra
Sometimes, malic acid, tartaric acid, CA and succinic acid were
amount of water is usually added in order to permit the complete
diffusion of acetone. Ultimately, centrifugation is used to separate used as a cross-linking agent for the preparation of CSNPs. The
NPs [61]. This method is schematically shown in Figure 9. carboxylic groups of di and tricarboxylic acids undergo chemical
cross-linking with the amino groups of CS, as a result, NPs forma-
tion take place [79]. Sodium sulphate is another important cross-
Cross linker of CSNPs linking agent used for the preparation of CSNPs.The sulphate
anion of sodium sulphate undergo crosslinking with the positively
Aldehyde
charged functional groups of CS [80].
Due to low-cost production, great fixation and the formation of a
thick cationic emulsion, glutaraldehyde is used as a versatile cross-
linker [62,63]. From the mechanistic point, a covalent crosslinking Physicochemical behaviour of CSNPs
takes place between the aldehyde groups of glutaraldehyde and Drug loading & drug release
the amino groups of CS, as a result, Nps formation takes place
[48]. Overt toxicity and balanced drug integrity are the drawbacks Drugs were loaded into CSNPs may be either incorporated the
of glutaraldehyde [64]. As per the report, the particle size has also drug at the time of NPs production or absorbed the drug after
been found to vary with the amount of glutaraldehyde [52]. the formation of NPs by incubating them in the drug solution.
Natural occurring a,b-unsaturated aldehyde, cinnamaldehyde is Diffusion and desorption are involved in the drug release from
another cross-linker used to prepared CSNPs [65]. Baicalein-loaded CSNPs [81]. The protein release from CSNPs may be due to three
CSNPs were successfully prepared by chemical cross-linking with predominant mechanisms including desorption, diffusion and
cinnamaldehyde as a cross-linking agent, by cross-linking method release due to polymer erosion of CSNPs [82]. Many factors were
in a W/O emulsion system [66]. Vanillin (3-methoxy-4-hydroxyben- found to influence the drug release rate from CSNPs including
zaldehyde) is another important phenolic aldehyde is used to pre- solubility, diffusion and biodegradation of the matrix materials,
pare CSNPs [67]. From the mechanistic point, chemical the choice of polymer matrices, the drug loading capacity and
crosslinking takes place between the aldehyde groups of vanillin size of NPs [1]. The equation which is used to describe drug
and the amino groups of CS, as a result, NPs formation take release mechanism
place [68].
Mt
¼ ktn ;
M1
Tripolyphosphate Mt
where M1 is equal to the drug release in the fraction at time t, k
TPP is a non-toxic, polyanion crosslinker. It has multivalent anions is equal to the constant which signify the macromolecular poly-
in its structure [69]. The amino groups of CS undergo reversible mer system properties.The n value is equal to the analysis of drug
physical cross-linking through electrostatic interaction with the release mechanism from the drug loaded NPs [83,84].
negatively charged TPP molecules, as a result, NPs formation takes
place [70,71]. Several advantages have been progressively indi-
Particle size &surface charge
cated for TPP, including the reversible physical cross-linking by
electrostatic interactionmakes it possible to prevent the toxicity of Most CSNPs prepared by the above methods have recorded mean
reagents [72]. sizes ranging from 100 to 400 nm. Particle size is one of the most
important features related to obtaining optimal in vitro efficacy of
CSNPs [85,86]. Particle size and its distribution were affected by
Genipin encapsulation efficiency and zeta potential [87]. Decrease the par-
Natural crosslinker genipin is obtained from gardenia fruit that ticles size could increase the specific surface area and the ratio of
has been utilised in herbal medicine and fabrication of food dyes. surface to volume, which could increase the dissolution and thus
The cytotoxicity of genipin is approximately 10,000 times less than increase the bioavailability of poorly water-soluble drugs [88].
glutaraldehyde [73,74]. Firstly, Song et al. [75] proposed the mech- Molecular weight and degree of deacetylation of CS are factors
anism of proteins crosslinking reaction by genipin. Kumar et al. found to affect particle size and surface charge [89]. Particle size
[76] synthesised ciprofloxacin-loaded genipin cross-linked CS/hep- and zeta potential are important factors which may influence the
arin nanoparticles (CIPRO-GP-CS/Hep NPs). They prepared these antifungal activity of NPs [90].
JOURNAL OF DRUG TARGETING 383

Zeta potential/surface charge is an important parameter of NPs [105]. Due to the mucoadhesive property, CS has been used
for the determination of their interaction in vivo with the nega- extensively as a carrier for mucosal drug delivery [106–108].
tively charged tumour cell membrane, stability, mucoadhesives Baltzley et al. [109] studied the probable use of CSNPs as an intra-
and permeation enhancing effect [91–93]. Zeta potential has the nasal delivery system to amplify olanzapine systemic bioavailabil-
greater influence on the particle stability in suspension. The less ity. They prepared these NPs through ionotropic gelation method.
zeta potential, the less likely the suspension to be stable [94]. They studied particle size, drug loading and in vitro release. Al-
Ghananeem et al. [110] studied the possible use of CSNPs as an
intranasal delivery system to amplify didanosine systemic and
Stability brain targeting efficiency. They prepared these NPs through iono-
The stability of NPs is an important factor in the pharmaceutical tropic gelation method and they studied prepared NPs size, drug
field until the loss of therapeutic efficacy [95]. Agglomerations of loading and in vitro release. Thus, both the intranasal route of
particles, bridging flocculation, coagulation are the predominant administration and formulation of didanosine in CSNPs aug-
factors for the physical instability of NPs [96]. Chemical stability of mented the delivery of didanosine to CSF and brain.
NPs depends on the following conditions like temperature, pH of
the medium, composition of the formulation, type of polymer Cancer drug delivery
used and the molecular weight of the polymer used in the formu-
lation. Most of the drugs sensitive to pH, hence pH should be Due to compatibility and biodegradability of CS, CSNPs are an
optimised to control the leakage of the drug [51]. CS chains pos- emerging field in the drug delivery of cancer [111]. CSNPs have
sess glucosamine groups that may be deprotonated if the pH been largely used for the delivery of anti-cancer drugs including
increases [97]. It was recorded that CSNPs prepared using sodium methotrexate [112], 5-flourouracil [113,114], cysplatin [115], epiru-
TPP and glycidoxy propyl trimethoxysilane cross-linking were pH bicin [116], curcumin [117,118], mitomycin C [119], docetaxel
sensitive [98]. Other research demonstrated that camptothecin- [120], doxorubicin (DOX) [49,121], paclitaxel [122] and tamoxifen
loaded N-isopropyl acrylamide-CSNPs were sensitive to tumour pH [123], focussing to augment anti-tumour efficacy, control release
when the charge ratio of N-isopropylacrylamide: CS was 4:1 [99]. and direct the drugs to the tumour thus reducing the toxicity.
In those studies, CSNPs were developed for: (i) to release 50%
of the methotrexate loaded in 48 h [112], (ii) to sustain release of
Cytotoxicity study& cellular uptake 5FU compared with the 5FU solution [113], (iii) to investigate NPs
were able to release the drug cysplatin in a sustained manner for
Cytotoxicity of CSNPs has been carried out to determine cell via-
a prolonged period of time [115], (iv) entrapment of epirubicin
bility. Cell viability and cell cytotoxicity have been carried out by a
into cholesterol-modified CS and show pH-dependent release of
simple non-radioactive colorimetric MTT (3–(4,5-dimethylthiazol-2-
drug in vitro [116], (v) encapsulation of doxorubicin in CS-based
yl)-2, 5 diphenyltetrazolium bromide) assay [100]. Corsi et al. [101]
NPs could be released into the cells in its active form [121], (vi)
prepared CS–DNA NPs and assessed the effect of prepared NPs
encapsulation of mitomycin C as intravesical chemotherapeutic
cell viability on human osteosarcoma cells, MSCs and HEK293,
agent and selectively incorporate NPs containing molecules in the
compared to a commercial lipid, Lipofect AMINETM2000 (LF). They
bladder cancer cell line, but not for normal bladder epithelial cells
measured the cell viability by MTT assay. In another study,
[119], (vii) the reduction of drug toxicity compared with free drug
Mansouri et al. [102] synthesised FA–CS–DNA NPs and evaluated
and reduce tumour volume in mice [120], (viii) carrier/vehicle/pro-
their effect on cell viability compared to LifofectAMINETM2000 (LF),
drug of cancer therapy [117,122,124–126], (ix) encapsulation of
a commercially available lipid vector [102]. The cytotoxic activities
dextran–doxorubicin conjugate in CSNPs [49], (x) to control the
of the CSNPs and copper (II)-loaded CSNPs was investigated by Qi
release of drug and to increase tamoxifen chemotherapeutic effi-
et al. [103] and a relationship between physiochemical properties ciency [123], (xi) chemotherapy of breast cancer in which the side
and activity is suggested. They used different cell lines for this effects of traditional chemo treatment could be reduced [114].
study. That author suggested that CSNPs and copper (II)-loaded
CSNPs showed much higher cytotoxicity toward BEL7402, BGC823
and Colo320 tumour cells while showing a slight effect on L-02 Anti-microbial drug delivery
human normal liver cells.
Efficient cellular uptake is an important parameter for NPs. Nowadays, CSNPs have been extensively utilised for drug delivery
applications in various infectious diseases. The functional groups
Confocal microscopic analysis is used to check the uptake of
help to aim loaded drugs to the site of infection. Slow biodegrad-
CSNPs into the cell nucleus [100] after 4 h exposure with 1% w/v
ation of CS permits controlled and sustained release of loaded
of CSNPs. Necrotic or autophagic cell death, possibly caused by
moieties, reduces the dosing frequency and suitable for raising
cell membrane damage is evoked by electron micrographs [104].
patient compliance in infectious diseases [127]. CSNPs have been
extensively used for the delivery of anti-microbial drugs including
Pharmaceutical applications of CSNPs cefazolin [128], rifampicin [129–131], daptomycin [132], ofloxacin
[133], amphoterecin B [134–136], vancomycin [137,138], ciprofloxa-
A specific administration route is highly essential for delivery of cin [139], amoxicillin [140], isoniazid [130,139] and tetracyc-
drugs to the site of its action [105]. A detail study of such admin- line [141].
istration to drug delivery is outlined below. In those studies, studies, CSNPs were developed for: (i) better
encapsulation efficiency (EE), good stability, excellent activity
against Klebseliapnominia, Paeroginosa and lactamase positive E.
Mucosal delivery
coli [128], (ii) better entrapment efficiency and good corneal per-
Nanocarriers have appeared as an effective strategy for mucosal meability [132], (iii) rapid bactericidal activity and reduced dose
drug delivery due to small steric obstruction and protection of frequency [129], (iv) better corneal permeation [133], (v) greater
freight therapeutics at both extracellular and intracellular range efficacy and better safety than commercial products [134], (vi)
384 S. NASKAR ET AL.

high entrapment efficiency, sustained release, prolong residence Vaccine delivery


time [135], (vii) to sustain release, low toxicity, preferential alveolar
CSNPs for parenteral and mucosal delivery of antigen vaccines
uptake and greater effectiveness [130], (viii) to sustain drug
have been developed [166–178]. CSNPs were widely apt for the
release [137], (ix) better internalisation, low dose and reducing modern vaccinology facilitated by the oral and nasal delivery of
dose frequency [142], (x) greater efficacy and complete removal of NPs with mucosal protective immune responses. The submicron
H. pylori [140], (xi) selective macrophage uptake and greater effi- size of NPs promotes their uptake up by Mcells, in mucosa-
cacy [131], (xii) prolonged residence time and better corneal per- associated lymphoid tissue, i.e. gut-associated, nasal-associated
meability [136], (xiii) to increase stability, targeted drug delivery and bronchus-associated lymphoid tissue and other targeting sites
and control the release of drug [138], (xiv) higher mucoadhesion of vigorous immunological responses [167]. CS-based DNA flu vac-
higher swelling [139], (xv) productive intracellular delivery and cine for intranasal administration was formulated [179]. Smitha
dose reduction [141]. et al. [180] developed O-carboxymethyl CSNPs encapsulated with
amidase. From this study, they concluded that the prepared NPs
can be used against S. aureus infections and CSNPs is the most
suitable candidate for the oral vaccine delivery system [180].
Colon-targeted drug delivery Pattani et al. [181] studied the immunological effects and mem-
Nowadays, CS is recognised as the polymer for oral site-specific brane interactions of CSNPs by using nitric oxide production, IL-6
drug delivery [143]. Ulcerative colitis, crohn’s disease, pseudomem- gene expression and lymphocyte proliferation involved in the
branous colitis and irritable bowel syndrome are the colonic dis- wound-healing process. Bivas-Benita et al. [182] studied the pul-
eases where CSNPs has been used in recent year [144,145]. CS is monary delivery of DNA vaccines against tuberculosis. From the
recognised as the polymer for colon-targeted delivery because it results, they concluded that pulmonary delivery of DNA vaccines
has a tendency to dissolve in acidic pH of the stomach and get may be a preferable delivery route compared to intramuscular
swollen in the intestinal pH [146]. immunisation.
As colon-targeted drug delivery, CSNPs were developed for: (i)
improving uptake in HT-29 cell and colorectal cancer [147], (ii) Transdermal drug delivery
releasing of 5-aminosalicylic acid from NPs was based on the ion-
exchange mechanism [148], (iii) sustaining in vitro drug release Transdermal route is generally considered as ‘patient-friendly’ due
[149], (iv) CS–DNA NPs were more stable in the upper regions of to the avoidance of gastrointestinal side effects which usually
the small intestine suggested that higher uptake rates may occur require many oral preparations [183]. In the recent study, Al-
in the duodenum compared with the ileum and the colon [150], Kassas et al. [184] prepared CSNPs dispersed in the mucoadhesive
(v) targeting drug delivery to colon tumour [151], (vi) in vivo fluor- gel for the transdermal delivery of propranolol hydrochloride. Zeta
escent endoscopic detection of colorectal cancer cells [152], (vii) potential was found to increase with increasing the concentration
the apoptosis was progressed on treatment with all-trans retinoic of CS. All prepared NPs showed 70% entrapment efficiency and
drug loading. From this study, they suggested that CSNPs dis-
acid bearing methoxy poly(ethylene glycol)-grafted CSNPs [153],
persed into gel enhanced the propranolol–HCl systemic bioavail-
(viii) 5-FU in hyaluronic acid-coupled CSNPs was about to 60-fold
ability. In another study, Hafner et al. [185] investigated the
more effective than free 5-FU on HT-29 cells [154].
potential CSNPs as colloidal nanosystem for transder-
mal melatonin.

Ocular delivery
Anti-inflammatory drug delivery
CS, hydrophilic natural biodegradable polymer improves stability,
CSNPs have been greatly used for the delivery of anti-inflamma-
precorneal retention and enhances interaction with eye mucosa.
tory drugs including zaltoprofen [186], hydrocortisone [97], ketoro-
Moreover, CS is recognised as the polymer suitable for ocular
lac tromethamine [188], tretinoin [189] aiming to increase anti-
drug delivery for the following reasons: non-toxic, low eye irrita- inflammatory activity, improved efficacy and high phys-
tion, the sustained release, mucoadhesive, in situ gelling, transfec- ical stability.
tion and permeation enhancing properties [155]. The In those studies, CSNPs were used for: (i) prolonging anti-
mucoadhesive polymer such as CS is able to form disulphide inflammatory activity [186], (ii) increasing anti-inflammatory activ-
bonds with mucus glycoproteins when found with the mucus gel ity and improving efficacy [187], (iii) showing anti-inflammatory
layer [156]. CSNPs have been extensively used for the delivery of activity in ocular diseases [188], (iv) greater physical stability, bet-
ocular drugs including CyA [34,157], FITC–BSA [158,159], indo- ter potential for topical delivery and augmenting therapeutic effi-
methacin [160,161], pilocarpine [162,163], pDNA [164] and pred- cacy [189].
nisolone [165].
In those studies, CSNPs were developed for: (i) increasing the
therapeutic index of clinically challenging drugs [34], (ii) demon- Miscellaneous drug delivery
strating biocompatibility of the CSNPs [158], (iii) increasing drug CSNPs have also been used as delivery systems for miscellaneous
levels in the anterior segment of the eye in connection with the classes of drug including anti-HIV (lamivudine [190], zidovudine
uncoated systems [160], (iv) increasing the concentration of drug [191]), anti-malarial (chloroquine [192], anti-tubercular (rifampicin
detected in the anterior segment and the vitreous versus indo- and isoniazid [130]), skeletal muscle relaxant (thiocolchicoside
methacin eye drops [161], (v) sustaining drug release and prolong- [193]) and oral hypoglycaemic (insulin [194]).
ing therapeutic effect of drug-loaded NPs [162,163], (vi) increasing In those studies, CSNPs were used for: (i) controlling drug
the drug levels detected in the aqueous humour versus those delivery applications [190], (ii) acting as drug delivery vehicle
observed with the commercial suspension [165]. against rodent parasite [192], (iii) sustaining the release of anti-
JOURNAL OF DRUG TARGETING 385

Table 1. Main characteristics of NPs based on TCS derivatives.


Drug Polymer Gelling agent Particle size (nm) Zeta potential (mV) References
Insulin CS–NAC TPP 140–210 19–31 [199]
Calcitonin Glycol CS–TGA TPP 230–330 21–27 [200]
OVA Thiolated TMC Thiolated HA 250–350 10–20 [201]
pSEAP CS–TGA None 212–113 4–8 [202]
None CS–TBA TPP 268 4–19 [203]

Table 2. Main characteristics of NPs based on amphiphilic CS.


Drug Polymer Particle size (nm) Zeta potential (mV) References
Camptothecin Glycol CS–5b–cholanic acid 250–350 Not reported [208]
None Glycol CS–5b–cholanic acid 230–310 10–11 [206]
Doxorubicin O-carboxymethyl CS-2,2I,(ethylene dioxy)-bis-(ethylamine)-folic acid 150–200 10–20 [215]
Griseofulvin/Cyclosporin A/Ranitidine Quaternary ammonium palmitoyl glycol CS 100–500 Not reported [212]
None Glycol CS–5b–cholanic acid 300–400 10 [209]
Doxorubicin Oleoyl–CS 250–350 Not reported [211]

Table 3. Main characteristics of NPs based on quaternised CS.


Drug Polymer Gelling agent Particle size (nm) Zeta potential (mV) References
Anti-neuroexcitation peptide TMC TPP 255 32 [224]
OVA TMC TPP 300 20 [223]
Insulin TMC Poly(c-glutamic acid) 100 30 [222]
Insulin TMC TPP 250 25 [225]
OVA TMC TPP 254–300 20–61 [226]
FITC–BSA TMC TPP Not reported Not reported [227]

tubercular drugs such as isoniazid and rifampicin to the Quaternised derivatives


lungs [130].
Trimethyl chitosan (TMC) being a quaternary derivative of CS has
shown better mucoadhesive, permeation enhancement, drug
Derivatives of CSNPs delivery and DNA delivery properties [216]. Examples of the above
quaternised CS derivative are the followings: N-TEC [183,217],
Thiolated chitosan N-diethyl methyl CS [218], N,N-dimethyl N-ethyl chitosan (DMEC)
Thiolated chitosan (TCS) is prepared by immobilisation of thiol [219] and N,N-diethyl N-methyl CS [220]. N-trimethylene chloride,
groups on the primary amino groups of CS backbone.TCS is a a quaternary derivative of CS has the better aqueous solubility,
new generation thiolated or thiolmer mucoadhesive polymer, pro- intestinal permeability and greater absorption over a wide pH
vides much better mucoadhesive properties than CS [195]. TCS range [221]. NPs loaded with insulin were developed from TMC by
NPs were expected to be themselves mucoadhesive, and hence, PEC method using poly(c-glutamic acid) as the polyelectrolyte
fit to prepare nanocarriers such as NPs for oral drug delivery complexing agent [222]. NPs loaded with OVA were developed
[196,197]. Examples of the above thiolated derivatives are the fol- from TMC using unmethylated CpG DNA as an adjuvant and a
lowing: CS–cysteine, CS–thiobutylamidine, CS–thioglycolic acid, crosslinker for nasal vaccination in mice [223]. Main characteristics
CS–polymethyl methacrylate [198], CS–sodium alginate [195] and of NPs based on quaternised CS shown in Table 3.
CS–glutathione conjugates. Main characteristics of NPs based on
TCS derivatives shown in Table 1.
CS nanocomplexes
CS formed nano-complex with an europium containing polyanion
Amphiphilic derivatives [CsEu6As6W63O218 (H2O)14(OH)4]25-(EuWAs) without the help of
Some amphiphilic derivatives such as glycol CS-5b-cholanic acid any crosslinker through ionotropic gelation method [228]. A few
conjugate [204–209], palmitoyl CS [210], oleoyl CS [211] were negatively charged active drugs, such as insulin or gene drugs,
developed by connecting hydrophobic structures to the CS back- when mixed with cationic CS derivatives in adequate proportions
bone. Other amphiphilic derivatives such as quaternary ammo- spontaneously formed nanoparticulate dispersions of insoluble
nium palmitoyl glycol CS [212] was developed from CS bearing complexes [202,225,229–232]. For the systemic delivery of SVN
fixed positive charges or linoleic acid-modified O-carboxymethyl siRNA, a hybrid CS nano-complex made of CS, protamine, lecithin
CS [213]/deoxycholic acid-modified N,O-carboxymethyl CS [214] and thiamine pyrophosphate was successfully developed. The tar-
were prepared from CS bearing fixed negative charges. Oleoyl CS, get gene silencing and cellular uptake efficiencies of the siRNA
hydrophobically modified CS derivative from which self-assembled nanocomplexes in prostate cancer cells (PC-3 cells) were meas-
NPs was obtained [211]. O-carboxymethyl CS, an amphiphilic CS ured. PC-3 tumour xenograft mouse model by near-infrared fluor-
derivative that would selfassemble into NPs when folic acid escence (NIRF) imaging and tumour growth monitoring was used
was conjugated with O-carboxymethyl CS via the bifunctional to assess in vivo tumour targetability and anti-tumour efficacy by
2,20 (ethylenedioxy)-bis-(ethylamine) [181]. Quaternary ammonium systemic administration [233]. Park et al. [234] prepared ionically
palmitoyl glycol CS is used to enhance absorption of hydrophobic crosslinked Ad/CS nanocomplexes for targeted cancer gene ther-
and hydrophilic drugs in rat orally [212]. Main characteristics of apy. They managed it by the process of electrospinning. Cheng
NPs based on amphiphilic CS shown in Table 2. et al. [235] have investigated the potential of DNA/CS
386 S. NASKAR ET AL.

nanocomplexes as a carrier for DOX. The in vivo biodistribution of References


FITC–CS and DNA/FITC–CS nanocomplexes after intravenous injec-
tion to the mice was evaluated by them. After 24 h post-injection, [1] Kumari A, Yadav SK, Yadav SC. Biodegradable polymeric
the DNA/FITC–CS nanocomplexes were accumulated into the liver nanoparticles based drug delivery systems. Colloids Surf B:
and kidney and remained at a relatively high stable level in blood, Biointerfaces. 2010;75:1–18.
while the fluorescence intensity of free FITC–CS decreased rapidly [2] Rampino A, Borgogna M, Blasi P, et al. Chitosan nanopar-
within 4 h post-injection. In vitro cytotoxicity test confirmed that ticles: preparation, size evolution and stability. Int J
DNA/CS–DOX conjugate exhibited cytotoxic effects on HeLa, Pharm. 2013;455:219–228.
[3] Diop M, Auberval N, Viciglio A, et al. Design, characterisa-
HepG2, QGY-7703 and L02 cells. In the study, Shah et al. [236]
tion, and bioefficiency of insulin-chitosan nanoparticles
three different nanoassemblies were made with CS. The
after stabilisation by freeze-drying or cross-linking. Int J
three nanoassemblies are CS–[NaP5W30O110]14 (CTS–P5W30),
Pharm. 2015;491:402–408.
Cs–[TeW6O24]6 (CTS–TeW6) and CS–[V10O28]6 (CTS–V10).
[4] Vimal S, Taju G, Nambi KSN, et al. Synthesis and character-
Zhao et al. [237] prepared TMC–cysteine conjugate (TMC–Cys)
ization of CS/TPP nanoparticles for oral delivery of gene in
nanocomplexes as a nonviral gene carrier.
fish. Aquaculture 2012;358–359:14–22.
[5] Tømmeraas K, Strand SP, Christensen BE, et al. Preparation
and characterization of branched chitosans. Carbohydr
Limitations Res. 2011;83:1558–1564.
[6] Chua BY, Kobaisi MAl, Zeng W, et al. Chitosan micropar-
Due to CS unique and versatile biological properties, it has been ticles and nanoparticles as biocompatible delivery vehicles
regarded as a useful polymer in the pharmaceutical area. for peptide and protein-based immunocontraceptive
Considerable research attempt has been done in order to develop vaccines. Mol Pharmaceutics. 2012;9:81–90.
safe and efficient CSNPs since 1994. However, poor stability of [7] Kean T, Thanou M. Biodegradation, biodistribution and
CSNPs regulates its practical applicability; thus, it has become a toxicity of chitosan. Adv Drug Deliv Rev. 2010;62:3–11.
significant challenge to form adequate shelf-life for CSNPs formu- [8] Kafetzopoulos D, Martinou A, Bouriotis V. Bioconversion of
lations. Stability can be improved by controlling the environmen- chitin to chitosan: purification and characterization of chi-
tal factors, temperature, launching a proper stabilising compound, tin deacetylase from Mucor rouxii. Proc Natl Acad Sci USA.
flourishing CS blends with another polymer and altering the CS 1993;90:2564–2568.
structure using chemical/ionic agents. The problem like, the solu- [9] Eijsink V, Hoell I, Vaaje-Kolstada G. Structure and function
bility of CS has to be resolved urgently. Only some hydrophilic of enzymes acting on chitin and chitosan. Biotechnol
drug can be encapsulated in unmodified CSNPs. Although modi- Genet Eng Rev. 2010;27:331–366.
fied CSNPs can encapsulate the hydrophobic drug. [10] Agrawal P, Strijkers GJ, Nicolay K. Chitosan-based systems
for molecular imaging. Adv Drug Deliv Rev. 2010;62:42–58.
[11] Nishimura K, Nishimura S, Seo H, et al. Macrophage activa-
tion with multiparous beads prepared from partially
Drawbacks and challenges deacetylated chitin. J Biomed Mater Res. 1986;20:
A nasal formulation of morphine (RylomineTM) based on CS is cur- 1359–1372.
rently in Phase 2 clinical trials (UK and EU) and Phase 3 clinical tri- [12] Kwon S, Park JH, Chung H, et al. Physicochemical charac-
als in the USA. We predict that when it comes to the market it teristics of self-assembled nanoparticles based on glycol
will approach the subject of similar products in the near future as chitosan bearing 5^a-cholanic acid. Langmuir 2003;19:
well as help in discerning any unforeseen outcome in humans 10188–10193.
[238]. Simple and mild preparation methods can be used to pre- [13] Yang Y, Wang S, Wang Y, et al. Advances in self-
pare CSNPs due to water-soluble property of CS which as an ideal assembled chitosan nanomaterials for drug delivery.
property for drug delivery carriers. In addition, CSNPs can be Biotechnol Adv. 2014;32:1301–1316.
[14] Ding F, Deng H, Du Y, et al. Emerging chitin and chitosan
administered through various route, especially non-invasive routes,
nanofibrous materials for biomedical applications.
i.e. peroral, nasal and ocular mucosa, which are preferable routes
Nanoscale 2014;6:9477–9493.
of administration and it acts as a good adjuvant in vaccine
[15] Hu X, Tang Y, Wang Q, et al. Rheological behaviour of chi-
delivery.Considerable attention has been directed to the applica-
tin in NaOH/urea aqueous solution. Carbohydr Polym.
tion of CSNPs.Bioavailability and absorption of drug enclosed into
2011;83:1128–1133.
CSNPs can be improved, so they can be used to deliver protein
[16] Wang Q, Zhang N, Hu X, et al. Chitosan/starch fibers and
drugs, gene drugs and can protect itself effectively from enzyme their properties for drug controlled release. Eur J Pharm
degradation in vivo. Biopharm. 2007;66:398–404.
[17] Wang Q, Jamal S, Detamore MS, et al. PLGA-chitosan/
PLGA-alginate nanoparticle blends as biodegradable col-
Disclosure statement loidal gels for seeding human umbilical cord mesenchy-
mal stem cells. J Biomed Mater Res A 2011;96A:520–527.
No potential conflict of interest was reported by the author(s). [18] Qun W, Yu-min D, Li h. F, et al. Structures and properties
of chitosan-starch-sodium benzoate blend films. J Wuhan
Univ (Nat Sci Ed) 2003;49:725–730.
Funding
[19] Ohya Y, Shiratani M, Kobayashi H, et al. Release behaviour
The authors wish to thank the University Grants Commission, of 5-fluorouracil from chitosan-gel nanospheres immobiliz-
Bahadur Shah Zafar Marg, New Delhi, India, for financial support. ing 5-fluorouracil coated with polysaccharides and their
JOURNAL OF DRUG TARGETING 387

cell specific cytotoxicity. J Macromol Sci A. 1994;31: [38] Songjiang Z, Lixiang W. Amyloid-beta associated with chi-
629–642. tosan nano-carrier has favorable immunogenicity and per-
[20] Kawashima Y, Handa T, Takenaka H, et al. Novel method meates the BBB. AAPS PharmSciTech. 2009;10:900–905.
for the preparation of controlled-release theophylline [39] Ngan LTK, Wang SL, Hiep M, et al. Preparation of chitosan
granules coated with a polyelectrolyte complex of sodium nanoparticles by spray drying, and their antibacterial activ-
polyphosphate-chitosan. J Pharm Sci. 1985;74:264–268. ity. Res Chem Intermed. 2014;40:2165–2175.
[21] Kawashima Y, Handa T, Kasai A, et al. The effects of thick- [40] Sinsuebpol C, Chatchawalsaisin J, Kulvanich P. Preparation
ness and hardness of the coating film on the drug release and in vivo absorption evaluation of spray dried powders
rate of theophylline granules coated with chitosan-sodium containing salmon calcitonin loaded chitosan nanopar-
tripolyphosphate complex. Chem Pharm Bull. 1985;33: ticles for pulmonary delivery. Drug Des Devel Ther.
2469–2474. 2013;7:861–873.
[22] Fernandez-Urrusuno R, Cavlo P, Remunan LC, et al. [41] Li FQ, Ji RR, Chen X, et al. Cetirizine dihydrochloride
Enhancement of nasal absorption of insulin using chitosan loaded microparticles design using ionotropic cross-linked
nanoparticles. Pharm Res 1999;16:1576–1581. chitosan nanoparticles by spray drying method. Arch
[23] Pan Y, Li Y, Zhao H, et al. Bioadhesive polysaccharide in Pharm Res. 2010;33:1967–1973.
protein delivery system: chitosan nanoparticles improve [42] Mehrotra A, Nagarwal RC, Pandit JK. Fabrication of lomus-
the intestinal absorption of insulin in vivo. Int J Pharm. tine loaded chitosan nanoparticles by spray drying and
2002;249:139–147. in vitro cytostatic activity on human lungcancer cell line
[24] Aydin RST, Pulat M. 5-Fluorouracil encapsulated chitosan L132. J Nanomed Nanotechnol. 2010;1:1–7.
nanoparticles for pH-stimulated drug delivery: evaluation [43] Tokumitsu H, Ichikawa H, Fukumori Y. Chitosan-
of controlled release kinetics. J Nanomater. 2012;2012:1. gadopentetic acid complex nanoparticles for gadolinium
[25] Doustgani A, Faraahani EV, Imani M. Preparation of chito- neutron-capture therapy of cancer: preparation by novel
san nanoparticles loaded bydexamethasone sodium phos- emulsion-droplet coalescence technique and characteriza-
phate. Iran J Pharm Res 2008;4:111–114. tion. Pharm Res. 1999;16:1830–1835.
[26] Leelapornpisid P, Leesawat P, Natakarnkitkul S, et al. [44] Reddy YD, Dhachinamoorthi D, Chandra SKB. Formulation
Application of chitosan for preparation of arbutin nano-
and in vitro evaluation of antineoplastic drug loaded
particles as skin whitening. JMMM 2010;20:101–105.
nanoparticles as drug delivery system. Afr J Pharm
[27] Saha P, Goyal AK, Rath G. Formulation and evaluation of
Pharmacol. 2013;7:1592–1604.
chitosan-based ampicillin trihydrate nanoparticles. Trop J
[45] Luque-Alcaraz AG, Lizardi-Mendoza J, Goycoolea FM, et al.
Pharm Res. 2010;9:483–488.
Preparation of chitosan nanoparticles by nanoprecipitation
[28] Rajendran NN, Natrajan R, Kumar RS, et al. Acyclovir-
and their ability as a drug nanocarrier. RSC Adv.
loaded chitosan nanoparticles for ocular delivery. Asian J
2016;6:59250–59256.
Pharm. 2010;4:220–226.
[46] Melo EP, Aires-Barros MR, Cabra JMS. Reverse micelles and
[29] Motwani SK, Chopra S, Talegaonkar S, et al. Chitosan-
protein biotechnology. Biotechnol Annu Rev. 2001;7:
Sodium alginate nanoparticles as submicroscopic reser-
87–129.
voirs for ocular delivery: formulation, optimization and
[47] Pileni MP. Reverse micelles used as templates: a new
in vitro characterization. Eur J Pharm Biopharm 2008;68:
understanding in nanocrystal growth. J Exp Nanosci.
513–525.
[30] Senthil V, Kumar RS, Nagaraju CVV, et al. Design and 2006;1:13–27.
development of hydrogel nanoparticles for mercaptopur- [48] Banerjee T, Mitra S, Singh AK, et al. Preparation, character-
ine. J Adv Pharm Tech Res. 2010;1:334–337. ization and biodistribution of ultrafine chitosan
[31] Nanjwade BK, Singh J, Parikh KA, et al. Preparation and nanoparticles. Int J Pharm. 2002;243:93–105.
evaluation of carboplatin biodegradable polymeric nano- [49] Mitra S, Gaur U, Ghosh PC, et al. Tumour targeted delivery
particles. Int J Pharm. 2010;385:176–180. of encapsulated dextran-doxorubicin conjugate using chi-
[32] Meng J, Sturgis TF, Youan BC. Engineering tenofovir tosan nanoparticles as carrier. J Control Release. 2001;74:
loaded chitosan nanoparticles tomaximize microbicide 317–323.
mucoadhesion. Eur J Pharm Sci. 2011;44:57–67. [50] Kafshgari MH, Khorram M, Mansouri M, et al. Preparation
[33] Alam S, Khan ZI, Mustafa G, et al. Development and evalu- of alginate and chitosan nanoparticles using a new
ation of thymoquinone encapsulated chitosan nanopar- reverse micellar system. Iran Polym J. 2012;21:99–107.
ticles for nose-to-brain targeting: a pharmacoscintigraphic [51] Alonso MJ. Nanoparticulate drug carrier technology. In:
study. Int J Nanomed. 2012;7:5705–5718. Cohen S, Bernstein H, editors. Microparticulate systems for
[34] De Campos AM, Sanchez A, Alonso MJ. Chitosan nanopar- the delivery of proteins and vaccines. New York: Marcel
ticles: a new vehicle for the improvement of the delivery Dekker; 1996. p. 203–242.
of drugs to the ocular surface. Application to cyclosporin [52] Janes KA, Calvo P, Alonso MJ. Polysaccharide colloidal par-
A. Int J Pharm. 2001;224:159–168. ticles as delivery systems for macromolecules. Adv Drug
[35] Wu Y, Yang W, Wang C, et al. Chitosan nanoparticles as a Deliv Rev. 2001;47:83–97.
novel delivery system for ammonium glycyrrhizinate. Int J [53] Poncelet D. Microencapsulation: fundamentals, methods
Pharm. 2005;295:235–245. and applications. In: Blitz JP, Gun’ko VM, editors. Surface
[36] Bhattarai N, Ramay HR, Chou SH, et al. Chitosan and lactic chemistry in biomedical and environmental science.
acid-grafted chitosan nanoparticles as carriers for pro- Dordrecht: Springer; 2006. p. 23–34.
longed drug delivery. Int J Nanomed. 2006;1:181–187. [54] Mao HQ, Roy K, Troung-Le VL, et al. Chitosan-DNA nano-
[37] Grenha A. Chitosan nanoparticles: a survey of preparation particles as gene carriers: synthesis, characterization and
methods. J Drug Target. 2012;20:291–300. transfection efficiency. J Control Release. 2001;70:399–421.
388 S. NASKAR ET AL.

[55] Borges O, Borchard G, Verhoef JC, et al. Preparation of release of alfuzosin hydrochloride. Pharm Res. 2013;30:
coated nanoparticles for a new mucosal vaccine delivery 512–522.
system. Int J Pharm. 2005;299:155–166. [74] Elzoghby AO, Helmy MW, Samy WM, et al. Spray-dried
[56] Atyabi F, Talaie F, Dinarvand R. Thiolated chitosan nano- casein-based micelles as a vehicle for solubilization and
particles as an oral delivery system for amikacin: in vitro controlled delivery of flutamide: formulation, characteriza-
and ex vivo evaluations. J Nanosci Nanotechnol. 2009;9: tion, and in vivo pharmacokinetics. Eur J Pharm Biopharm.
4593–4603. 2013;84:487–496.
[57] Agnihotri SA, Aminabhavi TM. Chitosan nanoparticles for [75] Song F, Zhang LM, Yang C, et al. Genipin-crosslinked
prolonged delivery of timolol maleate. Drug Dev Ind casein hydrogels for controlled drug delivery. Int J Pharm.
Pharm. 2007;33:1254–1262. 2009;373:41–47.
[58] Hu Y, Jiang X, Ding Y, et al. Synthesis and characterization [76] Kumar GV, Su CH, Velusamy P. Ciprofloxacin loaded geni-
of chitosan-poly(acrylic acid) nanoparticles. Biomaterials pin cross-linked chitosan/heparin nanoparticles for drug
2002;23:3193–3201. delivery application. Mater Lett. 2016;180:119–122.
[59] Sajeesh S, Sharma CP. Cyclodextrin-insulin complex encap- [77] Ahmed TA, Aljaeid BM. Preparation, characterization, and
sulated polymethacrylic acid based nanoparticles for oral potential application of chitosan, chitosan derivatives,
insulin delivery. Int J Pharm. 2006;325:147–154. and chitosan metal nanoparticles in pharmaceutical drug
[60] Sajeesh S, Sharma CP. Novel pH responsive polymetha- delivery. Drug Des Dev Ther. 2016;10:483–507.
crylic acid-chitosan-polyethylene glycol nanoparticles for [78] Bagheri M, Younesi H, Hajati S, et al. Application of chito-
oral peptide delivery. J Biomed Mater Res. 2006;76B: san-citric acid nanoparticles for removal of chromium (VI).
298–305. Int J Biol Macromol. 2015;80:431–444.
[61] El-Shabouri MH. Positively charged nanoparticles for [79] Bodnar M, Hartmann JF, Borbely J. Preparation and
improving the oral bioavailability of cyclosporin-A. Int J characterization of chitosan-based nanoparticles.
Pharm. 2002;249:101–108. Biomacromolecules 2005;6:2521–2527.
[62] Grasianto S, Siswanta, Andriani Y, et al. Retracted-glutaral- [80] Mayyas MA, Remawi A. Properties of chitosan nanopar-
dehyde-crosslinked chitosan-pectin nanoparticles as a ticles formed using sulfate anions as crosslinking bridges.
potential carrier for curcumin delivery and its in vitro Am J Appl Sci. 2012;9:1091–1100.
release study. Int J Drug Deliver. 2015;7:167–173. [81] Ringe K, Walz C, Sabel B. Nanoparticle drug delivery to
[63] Zhao LM, Shi LE, Zhang ZL, et al. Preparation and applica- the brain. In: Nalwa HS, editor. Encyclopedia of nano-
tion of chitosan nanoparticles.and nanofibers. Braz J Chem science and nanotechnology, vol. 7. New York: American
Eng. 2011;28:353–362. Scientific Publishers; 2004.
[64] Agnihotri SA, Mallikarjuna NN, Aminabhavi TM. Recent [82] Gan Q, Wang T. Chitosan nanoparticle as protein delivery
advances on chitosan-based micro- and nanoparticles in carrier—systematic examination of fabrication conditions
drug delivery. J Control Release. 2004;100:5–28. for efficient loading and release. Colloids Surf B:
[65] Balaguer MP, Gomez-Estaca J, Gavara R, et al. Functional Biointerfaces. 2007;59:24–34.
properties of bioplastics made from wheat gliadins modi- [83] Peppas NA. Analysis of Fickian and non-Fickian drug
fied with cinnamaldehyde. J Agric Food Chem. 2011;59: release from polymers. Pharm Acta Helv. 1985;60:110–111.
6689–6695. [84] Peppas NA, Korsmeyer RW. Dynamically swelling hydro-
[66] Babu VN, Kannan S. Enhanced delivery of baicalein using gels in controlled release applications. In: Peppas NA,
cinnamaldehyde cross-linked chitosan nanoparticle induc- editor. Hydrogels in medicine and pharmacy. Boca Raton,
ing apoptosis. Int J Biol Macromol. 2012;51:1103–1108. FL: CRC Press; 1987. p. 103–135.
[67] Li PW, Wang G, Yang ZM, et al. Development of drug- [85] Panyam J, Labhasetwar V. Biodegradable nanoparticles for
loaded chitosan-vanillin nanoparticles and its cytotoxicity drug and gene delivery to cells and tissue. Adv Drug Deliv
against HT-29 cells. Drug Deliv. 2016;23:30–35. Rev. 2003;55:329–347.
[68] Wang G, Li PW, Peng Z, et al. Formulation of vanillin [86] Moghimi SM, Hunter AC, Murray JC. Long-circulating and
crosslinked chitosan nanoparticles and its characterization. target-specific nanoparticles: theory to practice. Pharmacol
AMR. 2011;335-336:474–477. Rev. 2001;53:283–318.
[69] Liu Z, Jiao Y, Wang Y, et al. Polysaccharides-based nano- [87] Nagarwal RC, Singh PN, Kant S, et al. Chitosan nanopar-
particles as drug delivery systems. Adv Drug Deliv Rev. ticles of 5-fluorouracil for ophthalmic delivery: character-
2008;60:1650–1662. ization, in-vitro and in-vivo study. Chem Pharm Bull.
[70] Konecsni K, Low NH, Nickerson MT. Chitosan-tripolyphos- 2011;59:272–278.
phate submicron particles as the carrier of entrapped [88] Kreuter J. Peroral administration of nanoparticles. Adv
rutin. Food Chem. 2012;134:1775–1779. Drug Deliv Rev. 1991;7:71–86.
[71] Servat-Medina L, Gonzalez-Go mez A, Reyes-Ortega F, et al. [89] Tiyaboonchai W. Chitosan nanoparticles. A promising sys-
Chitosan-tripolyphosphate nanoparticles as Arrabidaea tem for drug delivery. Naresuan Univ J. 2003;11:51–66.
chica standardized extract carrier: synthesis, characteriza- [90] Ing LY, Zin NM, Sarwar A, et al. Antifungal activity of chi-
tion, biocompatibility, and anti ulcerogenic activity. Int J tosan nanoparticles and correlation with their physical
Nanomed. 2015;10:3897–3909. properties. Int J Biomater. 2012;2012:1–9.
[72] Hu B, Pan C, Sun Y, et al. Optimization of fabrication [91] Gaserod O, Jolliffe IG, Hampson FC, et al. The enhance-
parameters to produce chitosan-tripolyphosphate nano- ment of the bioadhesive properties of calcium alginate
particles for delivery of tea catechins. J Agric Food Chem. gel beads bycoating with chitosan. Int J Pharm.
2008;56:7451–7458. 1998;175:237–246.
[73] Elzoghby AO, Samy WM, Elgindy NA. Novel spray-dried [92] Smith J, Wood E, Dornish M. Effect of chitosan on epithe-
genipin-crosslinked casein nanoparticles for prolonged lial cell tight junctions. Pharm Res. 2004;21:43–49.
JOURNAL OF DRUG TARGETING 389

[93] Dong Y, Feng SS. Methoxy poly(ethylene glycol)-poly(lac- methotrexate release in vitro. Colloids Surf B: Biointerfaces.
tide) (MPEG-PLA) nanoparticles for controlled delivery of 2008;61:125–131.
anticancer drugs. Biomaterials 2004;25:2843–2849. [113] Zheng Y, Yang W, Wang C, et al. Nanoparticles based on
[94] Mu L, Feng SS. Fabrication, characterization and in vitro the complex of chitosan and polyaspartic acid sodium salt:
release of paclitaxel (TaxolV) loaded poly (lactic-co-glycolic
R
preparation, characterization and the use for 5-fluorouracil
acid) microspheres prepared by spray drying technique delivery. Eur J Pharm Biopharm. 2007;67:621–631.
with lipid/cholesterol emulsifiers. J Control Release. [114] Anitha A, Chennazhi KP, Nair SV, et al. 5-Flourouracil
2001;76:239–254. loaded N,O-carboxymethyl chitosan nanoparticles as an
[95] Abdelwahed W, Degobert G, Stainmesse S, et al. Freeze- anticancer nanomedicine for breast cancer. J Biomed
drying of nanoparticles: formulation, process and storage Nanotechnol. 2012;8:29–42.
considerations. Adv Drug Deliv Rev. 2006;58:1688–1713. [115] Cha J, Lee WB, Park CR, et al. Preparation and character-
[96] Hunter RJ. Foundations of colloid science, vol. I. New York: ization of cisplatin-incorporated chitosan hydrogels, micro-
Oxford Science Publications; 1993. p. 489–491. particles, and nanoparticles. Macromol Res. 2006;14:
[97] Lopez-Leon T, Carvalho ELS, Seijo B, et al. Physicochemical 573–578.
characterization of chitosan nanoparticles: electrokinetic [116] Wang YS, Liu LR, Jiang Q, et al. Self-aggregated nanopar-
and stability behavior. J Colloid Interface Sci. 2005;283: ticles of cholesterol-modified chitosan conjugate as a
344–351. novel carrier of epirubicin. Eur Polym J. 2007;43:43–51.
[98] Pan AW, Wu BB, Wu JM. Chitosan nanoparticles cross [117] Anitha A, Maya S, Deepa N, et al. Curcumin-loaded N,O-
linked by glycidoxy propyl trimethoxy silane for pH trig- Carboxymethyl chitosan nanoparticles for cancer drug
gered release of protein. Chin Chem Lett. 2009;20:79–83. delivery. J Biomater Sci Polym Ed. 2012;23:1381–1400.
[99] Fan L, Wu H, Zhang H, et al. Novel super pH-sensitive [118] Anitha A, Deepagan VG, Divya Rani VV, et al. Preparation,
nanoparticles responsive totumor extracellular pH. characterization, in vitro drug release and biological stud-
Carbohydr Polym. 2008;73:390–400. ies of curcumin loaded dextran sulphate-chitosan nano-
[100] Hu B, Ting Y, Zen X, et al. Cellular uptake and cytotoxicity particles. Carbohydr Polym. 2011;84:1158–1164.
of chitosan-caseinophos phopeptides nanocomplexes [119] Bilensoy E, Sarisozen C, Esendag  li G, et al. Intravesical cat-
loaded with epigallo catechin gallate. Carbohydr Polym. ionic nanoparticles of chitosan and polycaprolactone for
2012;89:362–370. the delivery of mitomycin C to bladder tumors. Int J
[101] Corsi K, Chellat F, Yahia LHocine, et al. Mesenchymal stem Pharm. 2009;371:170–176.
cells, MG63 and HEK293 transfection using chitosan-DNA [120] Hwang IS, Kim HY, Kwon IC, et al. Tumor targetability and
nanoparticles. Biomaterials 2003;24:1255–1264. antitumor effect of docetaxel-loaded hydrophobically
[102] Mansouri S, Cuie Y, Winnik F, et al. Characterization of modified glycol chitosan nanoparticles. J Control Release.
folate-chitosan-DNA nanoparticles for gene therapy. 2008;128:23–31.
Biomaterials 2006;27:2060–2065. [121] Janes KA, Fresneau MP, Marazuela A, et al. Chitosan nano-
[103] Qi L, Xu Z, Jiang X, et al. Cytotoxic activities of chitosan particles as delivery systems for doxorubicin. J Control
nanoparticles and copper-loaded nanoparticles. Bioorg Release. 2001;73:255–267.
Med Chem Lett. 2005;15:1397–1399. [122] Li F, Li J, Wen X, et al. Anti-tumor activity of paclitaxel-
[104] Loh JW, Yeoh G, Saunders M, et al. Uptake and cytotox- loaded chitosan nanoparticles: an in vitro study. Mater Sci
icity of chitosan nanoparticles in human liver cells. Toxicol Eng C. 2009;29:2392–2397.
Appl Pharmacol. 2010;249:148–157. [123] Vivek R, Nipun Babu V, Thangam R, et al. pH-responsive
[105] Liu M, Zhang J, Shan W, et al. Developments of mucus drug delivery of chitosan nanoparticles as tamoxifen car-
penetrating nanoparticles. Asian J Pharmacol. 2015;10: riers for effective anti-tumor activity in breast cancer cells.
275–282. Colloids Surf B: Biointerfaces. 2013;111:117–123.
[106] Lavelle E. Targeted mucosal delivery of drugs and vac- [124] Zhang X, Zhang H, Yin L, et al. A pH-sensitive nanosystem
cines. Expert Opin Ther Pat. 2000;10:179–190. based on carboxymethyl chitosan for tumor-targeted
[107] Lubben IMV, Verhoef JC, Borchard G, et al. Chitosan for delivery of daunorubicin. J Biomed Nanotechnol. 2016;12:
mucosal vaccination. Adv Drug Deliv Rev. 200;52:139–144. 1688–1698.
[108] Vila A, Sanchez A, Tobıo M, et al. Design of biodegradable [125] Zheng H, Yin L, Zhang X, et al. Redox sensitive shell and
particles for protein delivery. J Control Release. core crosslinked hyaluronic acid nanocarriers for tumor-
2002;78:15–24. targeted drug delivery. J Biomed Nanotechnol. 2016;12:
[109] Baltzley S, Mohammad A, Malkawi AH, et al. Intranasal 1641–1653.
drug delivery of olanzapine-loaded chitosan nanoparticles. [126] Hu R, Zheng H, Cao J, et al. Synthesis and in vitro charac-
AAPS Pharm Sci Tech. 2014;15:1598–1602. terization of carboxymethyl chitosan-cba-doxorubicin con-
[110] Al-Ghananeem AM, Saeed H, Florence R, et al. Intranasal jugate nanoparticles as pH-sensitive drug delivery
drug delivery of didanosine-loaded chitosan nanoparticles systems. J Biomed Nanotechnol. 2017;13:1097–1105.
for brain targeting; an attractive route against infections [127] Rajitha P, Gopinath D, Biswas R, et al. Chitosan nanopar-
caused by aids viruses. J Drug Target. 2010;18:381–388. ticles in drug therapy of infectious and inflammatory dis-
[111] Prabaharan M. Chitosan-based nanoparticles for tumor- eases. Expert Opin Drug Deliv. 2016;13:1177–1194.
targeted drug delivery. Int J Biol Macromol. 2015;72: [128] Jamil B, Habib H, Abbasi S, et al. Cefazolin loaded chitosan
1313–1322. nanoparticles to cure multidrug resistant gram-negative
[112] Yang X, Zhang Q, Wang Y, et al. Self-aggregated nanopar- pathogens. Carbohydr Polym. 2015;78:1–33.
ticles from methoxy poly(ethylene glycol)-modified [129] Dube A, Reynolds JL, Law WC, et al. Multimodal nanopar-
chitosan: synthesis, characterization; aggregation and ticles that provide immune modulation and intracellular
390 S. NASKAR ET AL.

drug delivery for infectious diseases. Nanomed [145] Bagre AP, Jain K, Jain NK. Alginate coated chitosan core
Nanotechnol. 2014;10:831–838. shell nanoparticles for oral delivery of enoxaparin: in vitro
[130] Garg T, Rath G, Goyal AK. Inhalable chitosan nanoparticles and in vivo assessment. Int J Pharm. 2013;456:31–40.
as antitubercular drug carriers for an effective treatment [146] Tozaki H, Odoriba T, Okada N, et al. Chitosan capsules for
of tuberculosis. Artif Cells Nanomed Biotechnol. colon-specific drug delivery: enhanced localization of
2016;44:997–1001. 5-aminosalicylic acid in the large intestine accelerates
[131] Moretton MA, Chiappetta DA, Andrade F, et al. Hydrolyzed healing of TNBS-induced colitis in rats. J Control Release.
galactomannan modified nanoparticles and flower-like 2002;82:51–61.
polymeric micelles for the active targeting of rifampicin to [147] Li P, Wang Y, Zeng F, et al. Synthesis and characterization
macrophages. J Biomed Nanotechnol. 2013;9:1076–1087. of folate conjugated chitosan and cellular uptake of its
[132] Costa JR, Silva NC, Sarmento B, et al. Potential chitosan- nanoparticles in HT-29 cells. Carbohydr Res. 2011;346:
coated alginate nanoparticles for ocular delivery of dapto- 801–806.
mycin. Eur J Clin Microbiol Infect Dis. 2015;34:1255–1262. [148] Saboktakin MR, Tabatabaie RM, Maharramov A, et al.
[133] € undag
Ust€ -Okur N, Go€kçe EH, Bozbıyık D_I, et al. Preparation Synthesis and in vitro evaluation of carboxymethyl starch-
and in vitro-in vivo evaluation of ofloxacin loaded ophthal- chitosan nanoparticles as drug delivery system to the
mic nano structured lipid carriers modified withchitosan colon. Int J Biol Macromol. 2011;48:381–385.
oligosaccharide lactate for the treatment of bacterial kera- [149] Akhlaghi SP, Saremi S, Ostad SN, et al. Discriminated
titis. Eur J Pharm Sci. 2014;63:204–215. effects of thiolated chitosan-coated pMMA paclitaxel-
[134] Jain V, Gupta A, Pawar VK, et al. Chitosan-assisted loaded nanoparticles on different normal and cancer cell
immunotherapy for intervention of experimental leish- lines. Nanomed: Nanotechnol. 2010;6:689–697.
maniasis via amphotericin B-loaded solid lipid nanopar- [150] Kadiyala I, Loo Y, Roy K, et al. Transport of chitosan-DNA
ticles. Appl Biochem Biotechnol. 2014;174:1309–1330. nanoparticles in human intestinal M-cell model versus
[135] Zhou W, Wang Y, Jian J, et al. Self-aggregated nanoparticles normal intestinal enterocytes. Eur J Pharm Sci. 2010;39:
based on amphiphilic poly(lactic acid)-grafted-chitosan 103–109.
copolymer for ocular delivery of amphotericin B. Int J [151] Jain A, Jain SK, Ganesh N, et al. Design and development
of ligand-appended polysaccharidic nanoparticles for the
Nanomed. 2013;8:3715–3728.
[136] Chhonker YS, Prasad YD, Chandasana H, et al. delivery of oxaliplatin in colorectal cancer. Nanomed
Nanotechnol. 2010;6:179–190.
Amphotericin-B entrapped lecithin/chitosan nanoparticles
[152] Yang SJ, Shieh MJ, Lin FH, et al. Colorectal cancer cell
for prolonged ocular application. Int J Biol Macromol.
detection by 5-aminolaevulinic acid-loaded chitosan nano-
2015;72:1451–1458.
particles. Cancer Lett. 2009;273:210–220.
[137] Kong Z, Yu M, Cheng K, et al. Incorporation of chitosan
[153] Park JS, Koh YS, Bang JY, et al. Antitumor effect of all-
nanospheres into thin mineralized collagen coatings for
trans retinoic acid-encapsulated nanoparticles of methoxy
improving the antibacterial effect. Colloids Surf B.
poly (ethylene glycol)-conjugated chitosan against CT-26
Biointerfaces. 2013;111:536–541.
colon carcinoma in vitro. J Pharm Sci. 2008;97:4011–4019.
[138] Pornpattananangkul D, Zhang L, Olson S, et al. Bacterial
[154] Jain A, Jain SK. In vitro and cell uptake studies for target-
toxin-triggered drug release from gold nanoparticle-stabilized
ing of ligand anchored nanoparticles for colon tumors.
liposomes for the treatment of bacterial infection. J Am
Eur J Pharm Sci. 2008;35:404–416.
Chem Soc. 2011;133:4132–4139. [155] Kapanigowda UG, Nagaraja SH, Ramaiah B, et al. Improved
[139] Junise V, Saraswathi R. Development and characterization
intraocular bioavailability of ganciclovir by mucoadhesive
of inhaled chitosan nanoparticles loaded with isoniazid. polymer based ocular microspheres: development and
JPTRM 2014;2:159–170. simulation process in wistar rats. DARU J Pharm Sci.
[140] Lin YH, Tsai SC, Lai CH, et al. Genipin-cross-linked fucose- 2015;23:11.
chitosan/heparin nanoparticles for the eradication of [156] Werle M, Bernkop SA. Thiolated chitosans: useful exci-
Helicobacter pylori. Biomaterials 2013;34:4466–4479. pients for oral drug delivery. J Pharm Pharmacol.
[141] Maya S, Indulekha S, Sukhithasri V, et al. Efficacy of tetra- 2008;60:273–281.
cycline encapsulated O-carboxymethyl chitosan nanopar- [157] Yuan X, Li H, Yuan Y. Preparation of cholesterol-modified
ticles against intracellular infections of Staphylococcus chitosan self-aggregated nanoparticles for delivery of
aureus. Int J Biol Macromol. 2012;51:392–399. drugs to ocular surface. Carbohydr Polym. 2006;65:
[142] Gnanadhas DP, Thomas MB, Elango M, et al. Chitosan-dex- 337–345.
tran sulphate nanocapsule drug delivery system as an [158] De Salamanca AE, Diebold Y, Calonge M, et al. Chitosan
effective therapeutic against intra phagosomal pathogen nanoparticles as a potential drug delivery system for the
Salmonella. J Antimicrob Chemother. 2013;68:2576–2586. ocular surface: toxicity, uptake mechanism and in vivo tol-
[143] Cota-Arriola O, Cortez-Rocha MO, Burgos HA, et al. erance. Invest Ophthalmol Vis Sci. 2006;47:1416–1425.
Controlled release matrices and micro/nanoparticles of [159] Diebold Y, Jarrın M, Saez V, et al. Ocular drug delivery by
chitosan with antimicrobial potential: development of new liposome-chitosan nanoparticle complexes (LCS-NP).
strategies for microbial control in agriculture. J Sci Food Biomaterials 2007;28:1553–1564.
Agric 2013;93:1525–1536. [160] Calvo P, Vila-Jato JL, Alonso MJ. Evaluation of cationic
[144] Hua S, Marks E, Schneider JJ, et al. Advances in oral nano- polymer-coated nanocapsules as ocular drug carriers. Int J
delivery systems for colon targeted drug delivery in Pharm. 1997;153:41–50.
inflammatory bowel disease: selective targeting to dis- [161] Calvo P, Alonso MJ, Vila-Jato JL, et al. Improved ocular
eased versus healthy tissue. Nanomed Nanotechnol. bioavailability of indomethacin by novel ocular drug car-
2015;11:1117–1132. riers. J Pharm Pharmacol. 1996;48:1147–1152.
JOURNAL OF DRUG TARGETING 391

[162] Kao HJ, Lo YL, Lin HR, et al. Characterization of pilocar- peritoneal macrophages. Int J Nanoparticles 2010;3:
pine-loaded chitosan/Carbopol nanoparticles. J Pharm 179–191.
Pharmacol. 2006;58:179–186. [179] Xu W, Shen Y, Jiang Z, et al. Intranasal delivery of chito-
[163] Lin HR, Yu SP, Kuo CJ, et al. Pilocarpine-loaded chitosan- san-DNA vaccine generates mucosal SIgA and anti-
PAA nanosuspension for ophthalmic delivery. J Biomater CVB3protection. Vaccine 2004;22:3603–3612.
Sci Polym Ed. 2007;18:205–221. [180] Smitha KT, Sreelakshmi M, Nisha N, et al. Amidase encap-
[164] De la Fuente M, Seijo B, Alonso MJ. Bioadhesive hyalur- sulated O-carboxymethyl chitosan nanoparticles for vac-
onan/chitosan nanoparticles can transport genes across cine delivery. Int J Biol Macromol. 2014;63:154–157.
the ocular mucosa and transfect ocular tissue. Gene Ther. [181] Pattani A, Patravale VB, Panicker L, et al. Immunological
2008;15:668–676. effects and membrane interactions of chitosan nanopar-
[165] Qu X, Khutoryanskiy VV, Stewart A, et al. Carbohydrate- ticles. Mol Pharm. 2009;6:345–352.
based micelle clusters which enhance hydrophobic [182] Bivas-Benita M, Van Meijgaarden KE, Franke KLMC, et al.
drug bioavailability by up to 1 order of magnitude. Pulmonary delivery of chitosan-DNA nanoparticles enhan-
Biomacromolecules 2006;7:3452–3459. ces the immunogenicity of a DNA vaccine encoding
[166] Vila A, Sanchez A, Janes K, et al. Low molecular weight HLA-A0201-restricted T-cell epitopes of Mycobacterium
chitosan nanoparticles as new carriers for nasal vaccine tuberculosis. Vaccine 2004;22:1609–1615.
delivery in mice. Eur J Pharm Biopharm. 2004;57:123–131. [183] Tanner T, Marks R. Delivering drugs by the transdermal
[167] Illum L, Jabbal-Gill I, Hinchcliffe M, et al. Chitosan as a route: review and comment. Skin Res Technol. 2008;14:
novel nasal delivery system for vaccines. Adv Drug Deliv 249–260.
Rev. 2001;51:81–96. [184] Al-Kassas R, Wen J, E-M, Cheng A, et al. Transdermal deliv-
[168] Amidi M, Romeijn SG, Verhoef JC, et al. N-trimethyl chito- ery of propranolol hydrochloride through chitosan nano-
san (TMC) nanoparticles loaded with influenza subunit particles dispersed in mucoadhesive gel. Carbohydr
antigen for intranasal vaccination: biological properties Polym. 2016;153:176–186.
and immunogenicity in a mouse model. Vaccine [185] Hafner A, Lovric J, Pepic I, et al. Lecithin/chitosan nanopar-
2007;25:144–153. ticles for transdermal delivery of melatonin.
[169] Boonyo W, Junginger HE, Waranuch N, et al. Chitosan and
J Microencapsul. 2011;28:807–815.
trimethyl chitosan chloride (TMC) as adjuvants for induc-
[186] Shah HA, Patel RP. Statistical modeling of zaltoprofen
ing immune responses to ovalbumin in mice following
loaded biopolymeric nanoparticles: characterization and
nasal administration. J Control Release. 2007;121:168–175.
anti-inflammatory activity of nanoparticles loaded gel. Int
[170] Svirshchevskaya EV, Alekseeva LG, Reshetov PD, et al.
J Pharma Investig. 2015;5:20–27.
Mucoadjuvant properties of lipo- and glycoconjugated
[187] Katas H, Hussain Z, Ling TC. Chitosan Nanoparticles as a
derivatives of oligochitosans. Eur J Med Chem.
percutaneous drug delivery system for hydrocortisone.
2009;44:2030–2037.
J Nanomater. 2012;2012:1:1–11.
[171] Sayin B, Somavarapu S, Li XW, et al. Mono-N-carboxy-
[188] Fardet L, Flahault A, Kettaneh A, et al. Corticosteroid-
methyl chitosan (MCC) and N-trimethyl chitosan (TMC)
induced clinical adverse events: frequency, risk factors and
nanoparticles for non-invasive vaccine delivery. Int J
patient’s opinion. Br J Dermatol. 2007;157:142–148.
Pharm. 2008;363:139–148.
[189] Ridolfi DM, Marcato PD, Justo GZ, et al. Chitosan-solid
[172] Borges O, Cordeiro-da-Silva A, Tavares J, et al. Immune
response by nasal delivery of hepatitis B surface antigen lipid nanoparticles as carriers for topical delivery of tretin-
and codelivery of a CpG ODN in alginate coated chitosan oin. Colloids Surf B Biointerfaces. 2012;93:36–40.
nanoparticles. Eur J Pharm Biopharm. 2008;69:405–416. [190] Dev A, Binulal NS, Anitha A, et al. Preparation of poly(lac-
[173] Klas SD, Petrie CR, Warwood SJ, et al. A single immuniza- tic acid)/chitosan nanoparticles for anti-HIV drug delivery
tion with a dry powder anthrax vaccine protects rabbits applications. Carbohydr Polym. 2010;80:833–838.
against lethal aerosol challenge. Vaccine 2008;26: [191] Dahmane EM, Rhazi M, Taourirte M. Chitosan nanopar-
5494–5502. ticles as a new delivery system for the Anti-HIV drug
[174] Florindo HF, Pandit S, Lacerda L, et al. The enhancement Zidovudine. Bull Korean Chem Soc. 2013;34:1333–1338.
of the immune response against S. equi antigens through [192] Tripathy S, Das S, Chakraborty SP, et al. Synthesis, charac-
the intranasal administration of poly-epsilon-caprolactone- terization of chitosan-tripolyphosphate conjugated chloro-
based nanoparticles. Biomaterials 2009;30:879–891. quine nanoparticle and its in vivo anti-malarial efficacy
[175] Baudner BC, Giuliani MM, Verhoef JC, et al. The concomi- against rodent parasite: a dose and duration dependent
tant use of the LTK63 mucosal adjuvant and of chitosan- approach. Int J Pharm. 2012;434:292–305.
based delivery system enhances the immunogenicity and [193] Nanda RK, Patil SS, Navathar DA. Chiotsan nanoparticles
efficacy of intranasally administered vaccines. Vaccine loaded with Thiocolchicoside. Der Pharma Chemica
2003;21:3837–3844. 2012;4:1619–1625.
[176] Amidi M, Mastrobattista E, Jiskoot W, et al. Chitosan-based [194] Makhlof A, Tozuka Y, Takeuchi H. Design and evaluation
delivery systems for protein therapeutics and antigens. of novel pH-sensitive chitosan nanoparticles for oral insu-
Adv Drug Deliv Rev. 2010;62:59–82. lin delivery. Eur J Pharm Sci. 2011;42:445–451.
[177] Cui Z, Mumper RJ. Chitosan-based nanoparticles for top- [195] Zhu X, Su M, Tang S, et al. Synthesis of thiolated chitosan
ical genetic immunization. J Control Release. 2001;75: and preparation nanoparticles with sodium alginate for
409–419. ocular drug delivery. Mol Vis. 2012;18:1973–1982.
[178] Madrigal-Carballo S, Esquivel M, Sibaja M, et al. Protein- [196] Kast CE, Bernkop SA. Thiolated polymers-thiomers: devel-
loaded chitosan nanoparticles modulate uptake and anti- opment and in vitro evaluation of chitosan-thioglycolic
gen presentation of hen egg-white lysozyme by murine acid conjugates. Biomaterials 2001;22:2345–2352.
392 S. NASKAR ET AL.

[197] Leitner VM, Walker GF, Bernkop-Schn€ urch A. Thiolated pol- characterization and application in vitro. Colloids Surf B:
ymers: evidence for the formation of disulphide bonds Biointerface. 2009;69:178–182.
with mucus glycoproteins. Eur J Pharm Biopharm. [214] Jin Y, Song Y, Zhu X, et al. Goblet cell-targeting nanopar-
2003;56:207–214. ticles for oral insulin delivery and the influence of mucus
[198] Saremi S, Atyabi F, Akhlaghi SP, et al. Thiolated chitosan on insulin transport. Biomaterials 2012;33:1573–1582.
nanoparticles for enhancing oral absorption of docetaxel: [215] Sahu S, Mallick SK, Santra S, et al. In vitro evaluation of
preparation, in vitro and ex vivo evaluation. Int J folic acid modified carboxymethyl chitosan nanoparticles
Nanomed. 2011;6:119–128. loaded with doxorubicin for targeted delivery. J Mater Sci:
[199] Wang X, Zheng C, Wu Z, et al. Chitosan-NAC nanoparticles Mater Med. 2010;21:1587–1597.
as a vehicle for nasal absorption enhancement of insulin. J [216] Mourya VK, Inamdar NN. Trimethyl chitosan and its appli-
Biomed Mater Res Part B Appl Biomater. 2009;88:150–161. cations in drug delivery. J Mater Sci Mater Med.
[200] Makhlof A, Werle M, Tozuka Y, et al. Nanoparticles of gly- 2009;20:1057–1079.
col chitosan and its thiolated derivative significantly [217] Avadi MR, Zohuriaan-mehr MJ, Younessi P, et al.
improved the pulmonary delivery of calcitonin. Int J Optimized synthesis and characterization of N-triethylchi-
Pharm. 2010;397:92–95. tosan. J Bioact Compat Polym. 2003;18:469–479.
[201] Verheul RJ, Sl€utter B, Bal S, et al. Covalently stabilized tri- [218] Avadi MR, Jalali A, Sadeghi AMM, et al. Diethyl methyl chi-
methyl chitosan-hyaluronic acid nanoparticles for nasal tosan as an intestinal paracellular enhancer: ex vivo and
and intradermal vaccination. J Control Release. 2011;156: in vivo studies. Int J Pharm. 2005;293:83–89.
46–52. [219] Bayat A, Sadeghi AMM, Avadi MR, et al. Synthesis of N,N-
[202] Martien R, Loretz B, Sandbichler AM, et al. Thiolated chito- dimethyl N-ethyl chitosan as a carrier for oral delivery of
san nanoparticles: transfection study in the Caco-2 differ- peptide drugs. J Bioact Compat Polym. 2006;21:433–444.
entiated cell culture. Nanotechnology 2008;19:045101. [220] Avadi MR, Sadeghi AMM, Tahzibi A, et al. Diethylmethyl
[203] Bernkop-schn€ urch A, Weithaler A, Albrecht K, et al. chitosan as antimicrobial agent: synthesis, characterization
Thiomers: preparation and in vitro evaluation of a and antibacterial effects. Eur Polym J. 2004;40:1355–1361.
[221] Thanou M, Verhoef JC, Marbach P, et al. Intestinal absorp-
mucoadhesive nanoparticulate drug delivery system. Int J
tion of octreotide: N-trimethyl chitosanchloride (TMC)
Pharm. 2006;317:76–81.
ameliorates the permeability and absorption properties of
[204] Yoo HS, Lee JE, Chung H, et al. Self-assembled nanopar-
the somatostatin analogue in vitro and in vivo.
ticles containing hydrophobically modified glycol chitosan
J Pharmaceutical Sci. 2000;89:951–957.
for gene delivery. J Control Release. 2005;103:235–243.
[222] Mi FL, Wu YY, Lin YH, et al. Oral delivery of peptide drugs
[205] Kim JH, Kim Y-S, Kim S, et al. Hydrophobically modified
using nanoparticles self-assembled by poly(c-glutamic
glycol chitosan nanoparticles as carriers for paclitaxel.
acid) and a chitosan derivative functionalized by trimethy-
J Control Release. 2006;111:228–234.
lation. Bioconjugate Chem. 2008;19:1248–1255.
[206] Park K, Kim JH, Nam YS, et al. Effect of polymer molecular
[223] €tter B, Jiskoot W. Dual role of CpG as immune modula-
Slu
weight on the tumor targeting characteristics of self-
tor and physical crosslinker in ovalbumin loaded
assembled glycol chitosan nanoparticles. J Control
N-trimethyl chitosan (TMC) nanoparticles for nasal vaccin-
Release. 2007;122:305–314.
ation. J Control Release. 2010;148:117–121.
[207] Kim J-H, Kim Y-S, Park K, et al. Self-assembled glycol chito-
[224] Wang S, Jiang T, Ma M, et al. Preparation and evaluation
san nanoparticles for the sustained and prolonged deliv- of anti-neuroexcitation peptide (ANEP) loaded N-trimethyl
ery of antiangiogenic small peptide drugs in cancer chitosan chloride nanoparticles for brain-targeting. Int J
therapy. Biomaterials 2008;29:1920–1930. Pharm. 2010;386:249–255.
[208] Min KH, Park K, Kim YS, et al. Hydrophobically modified [225] Sadeghi AM, Dorkoosh FA, Avadi MR, et al. Preparation,
glycol chitosan nanoparticles encapsulated camptothecin characterization and antibacterial activities of chitosan,
enhance the drug stability and tumor targeting in cancer N-trimethyl chitosan(TMC) and N-diethylmethyl chitosan
therapy. J Control Release. 2008;127:208–208. (DEMC) nanoparticles loaded with insulin using both the
[209] Nam HY, Won SMK, Chung H, et al. Cellular uptake mech- ionotropic gelation and polyelectrolyte complexation
anism and intracellular fate of hydrophobically modified methods. Int J Pharm. 2008;355:299–306.
glycol chitosan nanoparticles. J Control Release. 2009;135: [226] Amidi M, Romeijn SG, Borchard G, et al. Preparation and
259–267. characterization of protein-loaded N-trimethyl chitosan
[210] Chen KJ, Chiu YL, Chen YM, et al. Intracellularly monitor- nanoparticles as nasal delivery system. J Control Release.
ing/imaging the release of doxorubicin from pH-responsive 2006;111:107–116.
nanoparticles using Fo € rster resonance energy transfer. [227] Chen F, Zhang ZR, Yuan F, et al. In vitro and in vivo study
Biomaterials 2011;32:2586–2592. of N-trimethyl chitosan nanoparticles for oral protein
[211] Zhang J, Chen XG, Li YY, et al. Self-assembled nanopar- delivery. Int J Pharm. 2008;349:226–233.
ticles based on hydrophobically modified chitosan as car- [228] Menon D, Thomas RT, Narayanan S, et al. A novel chito-
riers for doxorubicin. Nanomed: Nanotechnol, Biol Med. san/polyoxometalate nanocomplex for anti-cancer applica-
2007;3:258–265. tions. Carbohydr Polym. 2011;84:887–893.
[212] Siew A, Le H, Thiovolet M, et al. Enhanced oral absorption [229] Bayat A, Larijani B, Ahmadian S, et al. Preparation and
of hydrophobic and hydrophilic drugs using quaternary characterization of insulin nanoparticles using chitosan
ammonium palmitoylglycol chitosan nanoparticles. Mol and its quaternized derivatives. Nanomedicine 2008;4:
Pharmaceutics. 2012;9:14–28. 115–120.
[213] Tan YL, Liu CG. Self-aggregated nanoparticles from linoleic [230] Bayat A, Dorkoosh FA, Dehpour AR, et al. Nanoparticles of
acid modified carboxymethyl chitosan: Synthesis, quaternized chitosan derivatives as a carrier for colon
JOURNAL OF DRUG TARGETING 393

delivery of insulin: ex vivo and in vivo studies. Int J targeted cancer gene therapy. J Control Release. 2010;148:
Pharm. 2008;356:259–266. 75–82.
[231] Jintapattanakit A, Junyaprasert VB, Mao S, et al. Peroral [235] Cheng X, Zhang F, Zhou G, et al. DNA/chitosan nanocom-
delivery of insulin using chitosan derivatives: a compara- plex as a novel drug carrier for doxorubicin. Drug Deliv.
tive study of polyelectrolyte nanocomplexes and nanopar- 2009;16:135–144.
ticles. Int J Pharm. 2007;342:240–249. [236] Shah HS, Al-Oweini R, Haider A, et al. Cytotoxicity and
[232] Lee D, Zhang W, Shirley SA, et al. Thiolated chitosan/DNA enzyme inhibition studies of polyoxometalates and
nanocomplexes exhibit enhanced and sustained gene their chitosan nanoassemblies. Toxicol Reports. 2014;1:
delivery. Pharm Res. 2007;24:157–167. 341–352.
[233] Ki MH, Kim JE, Lee YN, et al. Chitosan-based hybrid nano- [237] Zhao X, Yin L, Ding J, et al. Thiolated trimethyl chitosan
complex for siRNA delivery and its application for cancer nanocomplexes as gene carriers with high in vitro and in vivo
therapy . Pharm Res. 2014;31:3323–3334. transfection efficiency. J Control Release. 2010;144:46–54.
[234] Park Y, Kang E, Kwon OJ, et al. Ionically crosslinked Ad/ [238] Bellich B, D’Agostino I, Semeraro S, et al. “The good, the
chitosan nanocomplexes processed by electrospinning for bad and the ugly” of chitosans. Mar Drugs 2016;14:99.

View publication stats

You might also like