Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

Review

Journal of
Biomedical Nanotechnology
Copyright © 2016 American Scientific Publishers
All rights reserved Vol. 12, 1585–1603, 2016
Printed in the United States of America www.aspbs.com/jbn

Functional Chitosan Nanoparticles in Cancer Treatment


Shaozhi Fu1 ∗ , Jiyi Xia2 , and Jingbo Wu1
1
Department of Oncology, The Affiliated Hospital of Luzhou Medical College, Luzhou Medical College, Luzhou 646000, P. R. China
2
Department of Science and Technology, Luzhou Medical College, Luzhou 646000, P. R. China

With exponential growth in nanotechnology and material chemistry, the application of a variety of nanomaterials in biomed-
ical field has attracted increasing interest, especially in cancer diagnosis and treatment. Nanoparticles have greatly
revolutionized the therapy pattern of cancer owing to their specific targeting ability, prolonged circulation time, enhanced
therapeutic efficacy, and decreased systemic side effects. Chitosan, a modified natural polysaccharide with excellent bio-
compatibility, has been intensively studied for use as carriers to deliver various drugs and genes in cancer treatment. This
review provides an insight into the advances in chitosan-based nanoparticles with a focus on its therapeutic potential in
cancer treatment.
KEYWORDS: Chitosan Nanoparticles, Anticancer Drug, Drug Carrier, Gene Delivery, Biodegradability, Biocompatibility, Toxicity,
Structure Modification, Positive Targeting, Active Targeting, Blood Circulation Time, Multi-Drug Resistance, Blood Brain Barrier, Cancer
Diagnosis, In Vivo Imaging, Photodynamic Therapy, Thermotherapy, Individualized Cancer Therapy.

Delivered by Ingenta to: Main CID is 80004805 (JPP)


IP: 146.185.205.29 On: Wed, 07 Dec 2016 04:41:09
CONTENTS Directly Publishers
Copyright: American Scientific Conjugated Chemotherapeutic Drugs with
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1585 Chitosan Backbone . . . . . . . . . . . . ........ . . . . . . . . 1597
Source, Structure, and Physicochemical Encapsulation of Metal Compounds . . ........ . . . . . . . . 1598
Properties of Chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1586 Encapsulation of Protein . . . . . . . . . ........ . . . . . . . . 1598
Source and Structure . . . . . . . . . . . . . . . . . . . . . . . . . . . 1586 Problems and Outlook . . . . . . . . . . . . ........ . . . . . . . . 1599
Physicochemical Properties . . . . . . . . . . . . . . . . . . . . . . . 1587 Acknowledgements . . . . . . . . . . . . ........ . . . . . . . . 1599
Modification of Chitosan Nanoparticles . . . . . . . . . . . . . . . . . 1588 References . . . . . . . . . . . . . . . . . . ........ . . . . . . . . 1599
Chitosan Nanoparticles Modified by
Carbon-Based Materials . . . . . . . . . . . . . . . . . . . . . . . . . 1589
Chitosan Nanoparticles Modified by Natural Polysaccharide . . 1589 INTRODUCTION
Chitosan Nanoparticles Modified by Chemically It is well established that cancer has become one of the
Synthesized Copolymer . . . . . . . . . . . . . . . . . . . . . . . . . 1590 most serious threat to human health. It is estimated that
Chitosan Nanoparticles Modified by Low Molecular
there will be 12 million cancer deaths worldwide in 2030.1
Weight Compounds . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1590
Chitosan Nanoparticles Modified by Peptide, Protein . . . . . . . 1591 Especially in China, as a developing country with a large
Functions of Chitosan Nanoparticles . . . . . . . . . . . . . . . . . . . 1592 population, cancer incidence and death rates keep rising
Enhancing Anti-Tumor Efficacy . . . . . . . . . . . . . . . . . . . . 1592 year by year due to environmental pollution. According
Improving Special Targeting Tumor Tissue . . . . . . . . . . . . . 1592 to an estimate, six persons are diagnosed with cancer
Prolonging Blood Circulation Time . . . . . . . . . . . . . . . . . . 1592
every passing minute. This serious situation has brought
Reversal of Multi-Drug Resistance . . . . . . . . . . . . . . . . . . 1593
Crossing Blood-Brain Barrier . . . . . . . . . . . . . . . . . . . . . . 1593 a heavy burden to the society and patients. Among exist-
Diagnosis, Detection and Imaging . . . . . . . . . . . . . . . . . . . 1594 ing cancer treatments, chemotherapy is by far the most
Photodynamic Therapy and Imaging . . . . . . . . . . . . . . . . . 1594 employed form of intervention, however, patients face
Thermotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1595 many severe problems caused by chemotherapeutic agents,
Gene Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1596
such as detrimental side effects, drug resistance and high
Chitosan Nanoparticles in Drug Delivery . . . . . . . . . . . . . . . . 1597
Encapsulation of Chemotherapeutic Agents Into Chitosan . . . . 1597
cost.
In the process of exploring novel cancer therapy,

thanks to the progress in material science and process
Author to whom correspondence should be addressed.
Email: shaozhifu513@163.com technology, nanodrug development has emerged as a
Received: 17 May 2015 promising approach to overcome the shortcomings of con-
Revised/Accepted: 7 September 2015 ventional chemotherapeutic agents. Researchers worldwide

J. Biomed. Nanotechnol. 2016, Vol. 12, No. 8 1550-7033/2016/12/1585/019 doi:10.1166/jbn.2016.2228 1585


Chitosan Nanoparticles in Cancer Treatment Fu et al.

are now combining nanotechnology with drug delivery acid) (PLA), poly(-caprolactone) (PCL) or their polyethy-
system (DDS) to achieve interesting and useful results. lene glycol (PEG)-copolymers are well studied and widely
So far many nanoformulations of anticancer drugs have used as drug carriers.3 Natural biopolymers such as chi-
been developed and studied in clinical trails. For instance, tosan, collagen, cellulose, and fibrin, are also investi-
Abraxane® and Genexol-PM, two nanoformulations of gated intensively in pharmaceutical field owing to their
paclitaxel (PTX) have been approved for clinical use by unique characteristics.4 5 In particular, chitosan nanopar-
Food Drug Administration (FDA) in the past decade.2 ticles (CSNPs) have drawn considerable attention as
Therapeutic agents and their carriers are the two basic anticancer drug delivery carriers because of their easy
components of nanoformulations. The antitumor effect is accessibility, excellent stability, low toxicity, and easy
achieved by carriers delivering drug selectively to the modification.6 Herein, we aim to review various aspects
tumor cells. Therefore, the role of carriers in DDS is vital. of chitosan-based nanoparticles for drug delivery in cancer
The carriers for loading anticancer drugs are commonly treatment.
composed of amphiphilic polymers or hydrophilic biopoly-
mers. Based on the source of the composition matrix, drug SOURCE, STRUCTURE, AND
carriers can be classified into two types: chemosynthetic PHYSICOCHEMICAL PROPERTIES
and natural. Among chemically synthesized polymers, OF CHITOSAN
polyesters, poly(ether-esters), polyurethanes, and poly- Source and Structure
carbonates, have received considerable attention. Specif- As a natural polysaccharide, chitosan is manufactured on
ically, poly(lactic-co-glycolic acid) (PLGA), poly(lactic a large scale by alkaline N -deacetylation of chitin in

Shaozhi Fu received his Ph.D. from the State Key Lab of Biotherapy, Sichuan Univer-
sity, China in Chemical Biology in 2012. After finishing his graduate work, he became
a research assistant professor in Luzhou Medical College. He has served as a core
member of the Radiobiological Center in the Department of Oncology at the Affiliated
Delivered
Hospital ofby Ingenta
Luzhou to: Main
Medical CID since
College is 80004805
2012. His(JPP)
research interests include targeted
IP: 146.185.205.29 On: Wed, 07 Dec 2016
drug delivery system, biomedical nanotechnology04:41:09
and degradable biomaterials.
Copyright: American Scientific Publishers

Jiyi Xia received his M.D. from Luzhou Medical College, China in Clinical Medicine
in 2011. Then he became an experimentalist in Medical Experimental Center of the
Affiliated Hospital of Luzhou Medical College. He has served as a core member of
the Research Center of Drug and Functional Food in Luzhou Medical College since
2012. Currently, He is a member of the Department of Science and Technology, Luzhou
Medical College. His research interests include molecular biology, especially in cancer-
specific protein.

Jingbo Wu is a professor at the department of Oncology at the Affiliated Hospital of


Luzhou Medical College, China. He received his bachelor’s degree from the Department
of clinical medicine in the Chongqing Medical University in 1984. And then, he served
as a clinician in the Affiliated Hospital of Luzhou Medical College. In 1995, he got his
M.D. in medical oncology from Sun Yat-Sen Medical University, China. He is currently
the director of the Department of Oncology at the Affiliated Hospital of Luzhou Medical
College. His main interests include multimodality treatment of cancer, nanodrugs and
natural anti-caner products.

1586 J. Biomed. Nanotechnol. 12, 1585–1603, 2016


Fu et al. Chitosan Nanoparticles in Cancer Treatment

commercial production. Chitin is an abundant biopolymer of cell monolayers as well as an increase in the paracellu-
isolated from the exoskeleton of crustaceans, such as crabs lar permeability.8 9
and shrimps.7 Deacetylation of chitin and protonation
of chitosan is shown in the Figure 1. The proportion Biodegradability
of the two repeating units (glucosamine and N -acetyl- Biodegradability is a key property of polymers which are
glucosamine units) determines the degree of deacetylation used as carriers in DDS and as scaffolds in tissue engi-
of the polymer. neering. It not only determines the application potential
and range of biomaterials used in clinic, but also con-
Physicochemical Properties trols the metabolic fate of these polymers in the body.
Water Solubility Chitosan, as one of the biodegradable biopolymers, not
The aqueous solubility of chitosan has a great effect on only contains abundant amino groups, but also possesses
its processing conditions and application range. Chitosan hydrolysable glycosidic bonds in its backbone. Therefore,
is insoluble in aqueous medium at neutral pH but is sol- chitosan can be degraded to nontoxic oligosaccharides of
uble in slightly acidic environment owing to the amine variable length by proteases, mainly lysozyme, in vivo.
groups on its backbone. However, the solubility of chi- Subsequently, the produced oligosaccharides can be incor-
tosan in neutral and basic pH media can be improved porated in metabolic pathways or excreted out.10 Its degra-
by quaternization to form trimethylammonium chitosan dation rate is related to its molecular weight, the degree
derivatives. Moreover, the molecular weight of chitosan of deacetylation (DD), material shape, size, and adminis-
also greatly influences its solubility and degradability. Chi- tration route. Generally, higher molecular weight chitosan
has a lower degradation rate. Similar trend is observed
tosans and its derivatives having lower molecular weights
with DD, the degradation rate decreases with an increase
and lower degrees of deacetylation exhibit greater solu-
8 in the DD.
bility and faster degradation. Owing to the presence of
Chitosan is often used as a scaffold or carrier of active
protonatable amine groups, chitosan is a positively charged
biomolecules in tissue engineering. For this purpose, it
polymer at acidic pH (Fig. 1). This unique characteris-
is necessary for chitosan to possess enough persistence
tic endows it the ability to interact electrostatically with
time to allow the growth and extension of newborn tis-
polyanions when chitosan is used as carriers for deliv-
sue. For various applications as a relevant candidate for
ery of drugs or genes. It has been confirmed
Delivered bythat chitosan
Ingenta to: Mainmissing
CID isor80004805
damaged (JPP)
tissue and organ, chitosan scaffolds
with positive charge binds to cellIP: 146.185.205.29
membranes resultingOn:
in a Wed,can
07 Dec 2016 04:41:09
be easily processed into different shapes by method-
decrease in the transepithelial electricalCopyright:
resistance American
(TEER) Scientific Publishers
ologies, such as hydrogels, foams or sponges, and fibrous
membranes. Based on the adopted processing methods and
inherent properties of chitosan matrix, these biomaterials
with various morphologies may undergo in vivo degrada-
tion in days to weeks. For example, a chitosan derivative
(N ,O-carboxymethyl chitosan, with molecular weight of
200 kDa and 92% of DD) hydrogel persisted for ∼5 days
when it was used as a wound healing dressing.11 In a
yet another case, a chitosan-gelatin tridimensional scaffold
was observed even after 5 weeks of implantation into tooth
sockets of rats.12
Degradation of chitosan is also greatly influenced by
the administration route. After oral administration, chi-
tosan is degraded chemically as well as enzymatically via
hydrolysis of glucosamine–glucosamine, glucosamine-N -
acetyl-glucosamine, and N -acetyl-glucosamine-N -acetyl-
glucosamine linkages of chitosan.10 It has been confirmed
that chitosan and its derivatives are excellent nasal absorp-
tion enhancers. It widely used in formulations such as
solutions, gels, powders, and nanoparticles to deliver a
large amount of drugs owing to their unique efficacy in
improving the nasal bioavailability. When the chitosan-
based drugs are administrated by nasal route, they may
undergo the enzymatic degradation in the nasal cavity
by many enzymes including cytochrome-P450 dependent
Figure 1. Chemical structure of chitin, chitosan, and proto- monooxygenase, aldehyde dehydrogenase, peptidases and
nated chitosan. proteases, etc.13

J. Biomed. Nanotechnol. 12, 1585–1603, 2016 1587


Chitosan Nanoparticles in Cancer Treatment Fu et al.

Toxicity
Among natural polymers, chitosan is widely regarded
as non-toxic, biocompatible, and biodegradable polysac-
charide. Several products based on chitosan have been
proved by the FDA for use in wound dressing. How-
ever, when chitosan and its derivatives are used to deliver
drugs and genes, the toxicity of nanosized chitosan parti-
cles should not be ignored because the nanoparticles may
enter systemic circulation from the gastrointestinal tract,
nasal cavity, or alveolar sacs, thereby causing various lev-
els of toxicity to the human body.10 14 Also, in order to
achieve a specific targeting therapy, structural modification
of chitosan is carried out. This may lead to some toxicity
depending on the toxicity of the reactant(s) used.
Some studies have reported the toxicity evaluation of
CSNPs. The toxicity level of CSNPs may be related to sev-
eral factors including the properties of the parent material
which was used to prepare the nanoparticles, particle-size,
and the interacting cell type.15 Huang et al. reported that
the binding affinity and cell uptake capacity of CSNPs
reduced with the decrease of molecular weight (Mw) and
DD of the parent polymer. Cell uptake fell by 26% when
Mw was decreased from 213,000 to 10,000 and by 41%
when DD was lowered from 88% to 46%. Additionally,
the uptake data was also influenced by the zeta potential
of the nanoparticles. Chitosan molecules and nanoparticles
showed comparable cytotoxicityDelivered
profiles against the A549
by Ingenta to: Main CID is 80004805 (JPP)
cells and exhibited similar IC50 IP: IC20 values.16 On: Wed, 07 Dec 2016 04:41:09
and146.185.205.29
Since liver is the main detoxifying Copyright:
organ in theAmerican
human Scientific Publishers
body, hepatic toxicity of CSNPs should be taken into
consideration. CSNPs are known to show certain neg-
ative effects on hepatic cells. A study carried out by
Qi et al. reported that the incubation of human hepato-
cellular carcinoma-derived cell line (BEL7402 cells) with Figure 2. Investigation of the mechanism of toxicity caused
by chitosan nanoparticles. (A) cellular apoptosis was
CSNPs at 40 nm for 24 and 48 h displayed cytotoxicity determined using acridine orange staining of chitosan
with IC50 values of 15.01 and 6.19 g/mL, respectively.17 nanoparticle-exposed embryos at 96 post-fertilization.
Loh et al. also evaluated the hepatotoxicity of CSNPs.15 (B) Flow cytometric analysis of intracellular reactive oxygen
The cell toxicity analysis results revealed that human liver species in the control group (left panel) and 5 mg/L chitosan
nanoparticle-treated group (right panel). (C) Western blot
cells could tolerate 0.5% w/v of CSNPs (18 ± 1 nm,
analysis of HSP70 in zebrafish embryos. Reproduced with
7.5 ± 1.0 mV in culture medium) for up to 4 h. When permission from [14], Y. L. Hu, et al., Toxicity evaluation
the concentrations of chitosan nanoparticle were above of biodegradable chitosan nanoparticles using a zebrafish
0.5% w/v, cell membrane integrity was compromised as embryo model. Int. J. Nanomed. 6, 3351 (2011). © 2011,
evidenced by leakage of alanine transaminase into the DovePress.
extracellular milieu. Additionally, CYP3A4, an enzyme
Therefore, chitosan toxicity must be taken into account
found predominantly in the liver and intestinal enterocytes,
when CSNPs are used in systemic drug delivery.
showed a dose-dependent increase in its activity. CYP3A4
is responsible for approximately 60% of CYP-mediated
drug metabolism in human. Hu et al. used a zebrafish MODIFICATION OF CHITOSAN
embryo model to investigate the toxicity of CSNPs.14 Their NANOPARTICLES
results indicated that with an increase in the concentration Molecular backbone of chitosan contains reactive amino
of CSNPs, embryos showed a decreased hatching rate and and hydroxyl groups that can be readily decorated with
increased mortality. The physiological stress in zebrafish various ligands, functional groups and moieties.18 After the
caused by the nanoparticles is also evident by increased appropriate modification, beneficial properties can be
cytotoxicity, high expression of reactive oxygen species, as obtained including enhanced solubility, biocompatibility,
well as overexpression of heat shock protein 70 (Fig. 2).14 active targeting, etc. (Table I).

1588 J. Biomed. Nanotechnol. 12, 1585–1603, 2016


Fu et al. Chitosan Nanoparticles in Cancer Treatment

Table I. Modified chitosan derivatives used in anti-cancer drug delivery.

Modifier(s) Loading drug/gene(s) Cancer/cells Functions Ref.

Hyaluronic acid MiR-34a/Doxorubicin Breast cancer Combination therapy [26]


Heparin CdtB Gastric cancer Enhancing therapy efficacy [24]
PLGA Survivin siRNA Bladder cancer Gene therapy [29]
PNIPAM 5-FU – Thermo- and pH-sensitive [30]
PAMAM Lysozyme L929 fibroblast cells Gene therapy [31]
PEG ATRA U87MG cells Inhibiting tumor cell migration [32]
PEG-PCL Paclitaxel/Rutin – Prolonging drug release [33]
PLA Anthraquinone HepG2 cells Enhancing antitumor activity [34]
Folic acid (FA) pEGFP-N1 HeLa, MCF-7 Gene therapy [21]
CuAA HeLa, MCF-7, Hep-G2 Targeted delivery [35]
5-ALA HT29 cells Photodynamic detection [86]
NaYF4: Yb/Er H460 lung cancer cells Molecular diagnosis [96]
Oleic acid Cy5.5/ION U87MG cells Optical/MR imaging [36]
D,L-Lactic acid BSA – Prolonged drug delivery [39]
Glycol Paclitaxel Breast cancer Tumor-targeting therapy [54]
siRNA MCF-7/ADR Overcoming multi-drug resistance [72]
AuNPs CT-26 colon cancer cells Tumor-targeting CT imaging [90]
PpIX HT29 cells Photodynamic imaging and therapy [94]
Paclitaxel B16F10 melanoma tumor Enhanced therapeutic efficacy [123]
Cisplatin SCC7 cell-derived tumor Enhanced anti-tumor efficacy [134]
RGD peptide B16F10 melanoma tumor Antiangiogenic therapy [150]
Glycyrrhetinic acid DOX HepG2 Liver-cancer targeting therapy [41]
5-FU SMMC-7721 cells Liver-cancer targeting therapy [42]
Galactose (GC) 5-FU SMMC-7721 cells Liver-cancer targeting therapy [44]
Herceptin Gemcitabine Pancreatic cancer Enhanced antiproliferative activity [49]
Silk fibroin Proteins HepG2/CCL-13 cells Liver-cancer targeting therapy [50]
RGD peptide NaYF4:Yb/Er U87-MG/MCF-7 cells Photodynamic therapy [51]

Notes: Abbreviation: Hyaluronic acid (HA); Polyamidoamine (PAMAM); Poly(N-isopropylacrylamide) (PNIPAM); Poly(lactic-co-glycolic acid) (PLGA);
Delivered
Poly(ethylene glycol) (PEG), Methoxy poly(ethylene by Ingenta to:
glycol)–poly(lactic acid)Main CID isPoly(ethylene
(mPEG-PLA); 80004805glycol)–poly(-caprolactone)
(JPP) (PEG-PCL); All-trans
IP: 146.185.205.29
retinoic acid (ATRA); Green fluorescent protein On: Wed, 07
(pEGFP-N1); Copper acetylacetonate Dec 5-aminolevulinic
(CuAA); 2016 04:41:09 acid (5-ALA); Iron oxide nanoparticles (ION);
Bovine serum albumin (BSA); Gold nanoparticles Copyright: American
(AuNPs); Protoporphyrin Scientific
IX (PpIX); Publishers
Doxorubicin (DOX).

Chitosan Nanoparticles Modified by MWCNT-CS-FA NPs can be used as gene delivery vectors
Carbon-Based Materials for cancer treatment.
Carbon nanotubes (CNT) are carbon cylinders composed
of benzene rings. Although their insolubility in water Chitosan Nanoparticles Modified by
and organic solvents may cause toxicity, they are being Natural Polysaccharide
applied in biomedical fields as carriers to deliver pro- Heparin is a biodegradable, biocompatible, and negatively
tein, drugs, and sensors for DNA detection, inter alia.19 charged polysaccharide with many carboxylic groups in its
For the purpose of improving water-solubility and multi- molecular structure. It is relatively stable in vitro but it
functionalization, they can be chemically modified by can be degraded by hydrolysis and in vivo enzymolysis.22
conjugation with entities such as proteins, peptides, and Thus, it can be used to stabilize functional nanoparti-
bipolymers. However, the use of organic solvents may cles via the ionic interaction between cationic chitosan
limit polymer functionalization. To address the problem, and anionic heparin. A study reported by Yuk et al.
Li et al. developed multi-walled carbon nanotube chitosan revealed that gold-deposited iron oxide NPs immobilized
nanoparticle (MWCNT CSNP) hybrids by an ionotropic into the glycol chitosan/heparin network showed enhanced
gelation process. The in situ synthesis technique was tumor-specific targeting (Fig. 3).23 Lai et al. used heparin-
conducted at extremely mild room temperature condi- modified chitosan NPs to load cytolethal distending toxin
tions without the use of any toxic solvents. The pre- (CdtB) for the treatment of gastric cancer. Like CdtB
pared MWCNT-CSNP hybrids improved the model protein itself, the CdtB-encapsulated nanoparticles could enhanc
immobilization efficiency 0.8 times and simultaneously cell-cycle arrest at G2/M leading to apoptosis. The mech-
decreased the cellular toxicity by ∼50% compared with anism for CdtB-encapsulated nanoparticles-induced cell
carboxylated MWCNT.20 MWCNTs with varying length death was mediated by ATM-dependent DNA damage
were also conjugated with chitosan-folic acid nanoparticles checkpoint responses.24
(CS-FA NPs) by ionotropic gelation process. The surface Hyaluronic acid (HA) is a biodegradable and lin-
functionalization improves the transfection efficiency and ear polysaccharide with high binding capacity to HA
decreases the cytotoxicity of MWCNTs.21 Therefore, the receptors (trans-membrane glycoprotein CD44). Therefore,

J. Biomed. Nanotechnol. 12, 1585–1603, 2016 1589


Chitosan Nanoparticles in Cancer Treatment Fu et al.

Figure 3. In vivo noninvasive NIR images and quantification analysis of the Cy5.5/composite NPs with or without heparin.
Reproduced with permission from [23], S. H. Yuk, et al., Glycol chitosan/heparin immobilized iron oxide nanoparticles with a
tumor-targeting characteristic for magnetic resonance imaging. Biomacromolecules 12, 2335 (2011). © 2011, American Chemical
Society.

HA modified DDS can specifically improve drug accu- and/or to the tumor site, thereby increasing the therapeutic
mulation in cancer cells over-expressing CD-44. For response.29
example, polyelectrolyte complex nanoparticles based on Poly(N -isopropylacrylamide) (PNIPAA) is a widely
hyaluronic acid/chitosan (HA/CS) showed potent cytotox- studied polymer with excellent thermosensitivity. The
icity and higher uptake efficiency in C6 cells after load- PNIPAA-modified CSNPs has the potential to exhibit
ing a water-insoluble curcuminoid.25 Deng et al. evidenced unique thermo-responsibility. Zhang et al. prepared self-
simultaneous co-delivery of chemotherapeutic agent dox- assembled chitosan-graft-poly(N -isopropylacrylamide)/
orubicin (DOX) and tumor suppressive
DeliveredmiRNA-34a
by Ingenta into
to: Maincarboxymethyl
CID is 80004805 cellulose
(JPP) nanoparticles to load
IP: 146.185.205.29
triple negative breast cancer cells by HA-CSNPs. ItOn: notWed, 07 Dec 2016
5-fluorouracil 04:41:09
(5-FU). The obtained polyelectrolyte com-
only improved the antitumor activity Copyright:
of DOX, butAmerican
it also Scientific Publishers were thermosensitive and had an
plex nanoparticles
suppressed tumor cells migration by targeting Notch-1 average diameter of about 200 nm.30
26
signaling. Besides, HA-CSNPs also have a great poten- Polyamidoamine dendrimers are a new type of synthetic
tial in ocular gene delivery.27 polymer characterized by a branched spherical shape and a
Apart from the above mentioned polysaccharides, other high density surface charge. Zhang et al. grafted carboxyl
polysaccharides such as alginate, starch, pectin, and car- group-poly(amidoamine) onto carboxymethyl chitosan for
boxymethyl cellulose are also used to fabricate polyelec- preparing core–shell nanoparticles.31 The self-assembled
28
trolyte complexes with chitosan for drug delivery. dendrimer nanoparticles did not display significant cyto-
toxicity in the range of concentrations below 3.16 mg/ml
Chitosan Nanoparticles Modified by Chemically and showed excellent properties as highly potent and non-
Synthesized Copolymer toxic intracellular carriers for protein delivery.
Many chemically synthesized copolymers like polyesters In addition to these, other polymers, such as polyethy-
and polyamides have been used directly as drug carriers lene glycol, methoxy poly(ethylene glycol)–poly(lactic
owing to their excellent biocompatibility and controllable acid) (mPEG-PLA), poly(ethylene glycol)–poly(-
degradability. They can also be used to improve the bio- caprolactone) (PEG-PCL), have also been grafted onto
logical and chemical properties of chitosan by chemical chitosan for fabricating various carrier nanoparticles.32–34
conjugation. For example, Chen et al. used paclitaxel and rutin as
Poly(lactic-co-glycolic acid) (PLGA) is a biocompati- model drugs for the evaluation of the controlled release
ble and degradable polymer that has been widely used to capacity of the chitosan-PEG-PCL nanoparticles.33 Wang
encapsulate and deliver various chemotherapeutic drugs, et al. prepared cationic biodegradable mPEG-PLA-CSNPs
siRNAs, DNAs, peptides, and proteins. Chitosan-modified as a small interfering ribonucleic acid (siRNA) delivery
34
PLGA nanoparticles can protect biologic agents from system for hepatitis B.
degradation during systemic circulation, thereby enhanc-
ing therapeutic efficacy. Martin et al. reported that Chitosan Nanoparticles Modified by Low
PLGA-chitosan (chitosan with a low molecular weight Molecular Weight Compounds
of 2.5 kDa) nanoparticles have the capacity to transport The compounds with low molecular weight offer
large amounts of surviving siRNA across the urothelium unique characteristics in modification of nanoparticles.

1590 J. Biomed. Nanotechnol. 12, 1585–1603, 2016


Fu et al. Chitosan Nanoparticles in Cancer Treatment

The modified NPs may exhibit targeting efficacy, lower chitosan nanoparticles can target the liver, and have sig-
toxicity, longer circulation time, etc. Folate, a low molec- nificantly inhibited tumor growth in an orthotropic liver
ular weight compound, is often used as a modifier owing cancer mouse model.42
to high-expression of folate receptor in many cancer In order to enhance liver-targeted drug delivery, galac-
cells. Folic acid can be covalently conjugated to chi- tosylated nanocarrier is another ideal option. Asialo-
tosan molecules via its gamma-carboxyl moiety. The glycoprotein receptors are expressed predominantly on
folic acid-tagged hydrophobic-modified chitosan NPs has hepatocytes for clearance of galactose-terminated gly-
been shown to load curcumin and successfully deliver coproteins, and have been identified on a continuous
the drug to folate receptor over-expressed cancer cells.35 human hepatoma cell line, HepG2.43 Therefore, the ther-
Oleic acid is often used to prepare magnetic iron oxide apeutic activity can be improved by the interactions
nanoparticles (Fe3 O4 NPs), and the oleic acid-decorated between the ligand and liver cancer cell receptors. Cheng
Fe3 O4 NPs are usually encapsulated in various carriers et al. used galactosylated chitosan (GC) to prepare 5-FU
for use as imaging probes to detect tumors. However, loaded nanoparticles (GC/5-FU). Analysis of apoptosis
the interface compatibility between Fe3 O4 NPs and carri- pathways indicated that GC/5-FU upregulates p53 expres-
ers can greatly influence the loading efficacy and imag- sion at both protein and mRNA levels. In vivo antitumor
ing quality. Oleic acid is used to modify the polymeric evaluation showed that the sustained release of GC/5-FU
carriers to improve the interface compatibility. After graft- nanoparticles was more effective at targeting hepatic can-
ing with oleic acid, oleoyl-chitosan self-assembles into cer cells than 5-FU monotherapy in the mouse orthotropic
core–shell structures in aqueous solution and provides liver cancer mouse model.44 Owing to its excellent active
36 targeting ability, galactosylated chitosan has been widely
the effective core compartment for loading Fe3 O4 NPs.
A high-performance nanoparticle is prepared by physi- used as nanocarriers for delivering various therapeutic
cally complexing succinate-modified chitosan with folic drugs and genes in cancer treatment.45–48
37
acid-modified chitosan. The resulting nanoparticles have
an average diameter and zeta potential of 110.0 nm and Chitosan Nanoparticles Modified by
18.6 mV, respectively. They are very stable in aqueous Peptide, Protein
suspension and are readily engulfed by oral cancer cells The bioactive molecules including peptides and proteins
via folate-receptor-mediated endocytosis. Therefore, this are also used to prepare functionalized chitosan NPs with
kind of chitosan nanoparticle is Delivered by Ingenta
an excellent vector to:
for Main CID isfunctions.
unique 80004805 (JPP)
IP: 146.185.205.29 On:
oral-specific delivery of 5-aminolevulinic acid for fluores- Wed, 07 Dec 2016 04:41:09
In order
Copyright: American Scientific to enhance selective delivery of anti-tumor
Publishers
cent endoscopic detection, and it can be used for pho- drugs to tumor sites, herceptin was conjugated with
todynamic detection of oral cancer. Jin et al. prepared gemcitabine-loaded CSNPs (HER2-Gem-CSNPs).49 The
N -octyl-O-sulfated chitosan (NOSC) with a viscosity aver- targeted NPs displayed marked cytotoxicity along with an
age molecular weight of 65–70 kDa for the prepara- enhanced S-phase arrest, leading to apoptosis in compari-
tion of PTX-loaded micelles.38 The PTX micelles showed son with free gemcitabine and unconjugated gemcitabine-
superior blood persistence, tumor accumulation, and ther- loaded nanoparticles due to higher cellular binding with
apeutic efficacy after intravenous injection into the tumor- eventual uptake and prolonged intracellular retention.
bearing mice. Thus, HER2-Gem-CSNPs are able to provide an efficient
D,L-Lactic acid has also been used as a modifier to dec- and targeted delivery of gemcitabine for pancreatic cancer
orate chitosan by grafting lactic acid onto amino groups treatment.
in chitosan. After incorporating the lactyl segment into Silk fibroin is an insoluble protein with low immune or
the chitosan backbone, the resulting nanoparticle showed inflammatory response and favorable biological response
a high protein encapsulation (96%) and a prolonged drug characteristics. Therefore, silk fibroin-modified CSNPs are
release rate (15%) over 4 weeks.39 Glycyrrhetinic acid very suitable for chemotherapeutic delivery in cancer treat-
(GA) is one of the main bioactive compounds of licorice ment because of their better stability, low toxicity, simple
and is used widely as medicine for the treatment of and mild preparation methods.50 Zhou et al. developed
many diseases. It is also used as a ligand to target liver a kind of upconversion nanoparticle (UCNP-Ppa-RGD)
because of the abundance of receptors for GA on the hep- by using a photosensitizer pyropheophorbide a (Ppa) and
atocyte membrane.40 Tian et al. used GA-modified sul- RGD peptide c (RGDyK) to co-modify chitosan and
fated chitosan (GA-SCS) to prepare doxorubicin-loaded wrap NaYF4:Yb/Er nanocrystals.51 The UCNP-Ppa-RGD
41
micelles (DOX/SA-SCS micelles). The IC50 of the pre- nanoparticles exhibited low dark toxicity and high photo-
pared micelles against liver cancer cells (HepG2 cells) was toxicity against cancer cells upon 980 nm laser irradiation
54.7 ng/mL, remarkably lower than that of the no-GA- at an appropriate dose indicating that they may be used
modified micelles. Moreover, the DOX/SA-SCS micelles for near-infrared photodynamic therapy with an improved
showed higher affinity for the liver cancer cells (HepG2 tumor targeting specificity.
cells) than for the normal liver cells (Chang liver cells). Besides the above mentioned peptides and proteins,
Another study reported that 5-FU-conjugated GA-modified other proteins have also been applied to modify chitosan

J. Biomed. Nanotechnol. 12, 1585–1603, 2016 1591


Chitosan Nanoparticles in Cancer Treatment Fu et al.

for preparing the nanoparticles with unique physicochem- delivery system. It has been intensively studied in various
ical properties. For example, lectin conjugated chitosan drug delivery systems. The nanoparticles based on galac-
nanoparticles showed fluorescence characteristics with a tosylated chitosan (GC) and 5-FU can be more effective at
potential to be used for disease detection and drug targeting hepatic cancer cells than 5-FU monotherapy in
delivery.52 the orthotropic liver cancer mouse model because Asialo-
glycoprotein receptor (ASGPR) found on membranes of
FUNCTIONS OF CHITOSAN hepatocytes shows specificity for glycoproteins.44 Gly-
NANOPARTICLES cyrrhetinic acid-modified sulfated chitosan (GA-SCTS)
Even though chitosan nanoformulations are seldom used micelles specifically target the liver cancer cells (HepG2
in cancer treatment up till now, multi-functional CSNPs cells) and display rapid and significant ability to target
are showing promise in personalized therapy. The nanopar- the liver in vivo.41 42 An aptamer conjugated hyaluro-
ticles may show improved anti-tumor efficacy and spe- nan/CSNPs were prepared and used as carriers for targeted
cific targeting ability due to modification of chitosan delivery of 5-FU by Ghasemi et al. The prepared NPs
nanocarriers or encapsulation of multiple therapeutic showed significantly higher targeting ability in (MUC1+
agents. human colorectal adenocarcinoma as compared with the
free drug.57
Enhancing Anti-Tumor Efficacy
In clinical treatment, many anticancer drugs with tra- Prolonging Blood Circulation Time
ditional formulation are being despised due to certain During the development of effective drug delivery
disadvantages such as poor water solubility, short cir- nanoparticles, one major obstacle is the rapid clearance
culation time in vivo, poor targeting and high side from blood. The ability of drug-loaded nanoparticles
effects.53 Nanomedicine holds a great potential in resolv- to circulate in the bloodstream for a prolonged period
ing these problems. Nanoparticles have been considered of time is often a prerequisite for successful targeted
to be effective carriers because they can stay unrec- delivery.58 59 They should evade the phagocytic uptake by
ognized during blood circulation, reduce the adverse reducing opsonization by blood proteins, hence increasing
reactions and increase the therapeutic efficacy.3 As one the bioavailability of the drug. The in vivo fate of nanopar-
of drug carriers, CSNPs mayDelivered by Ingenta
show enhanced to: Mainticles
antitu- CID isis 80004805 (JPP) on the chemical and physi-
mainly dependent
mor efficacy in cancer treatment IP:owing
146.185.205.29 On: Wed, 07 Dec 2016 04:41:09
to many unique cal properties of the NPs, including size, surface charge
characteristics.5 Water soluble chitosanCopyright: American
naonoparticles can Scientific
and
Publishers
surface chemistry. 60 61
In order to overcome these
improve water solubility of hydrophobic drugs.54 55 The obstacles, some shielding groups including PEG, polyvinyl
biodegradability of chitosan matrix may ensure that encap- alcohol, and polysaccharides are adsorbed or grafted on
sulated drugs are released in a controlled fashion.4 18 The the surface of NPs for masking the NPs because these
small-sized CSNPs can pass through biological barriers
groups or polymers can hinder the hydrophobic and elec-
in vivo and deliver drugs to the tumor site owing to
trostatic interactions that help plasma proteins bind to par-
the enhanced permeation and retention or active target-
ticle surface.62–64
ing ability.47 The specific targeting of modified CSNPs
For the purpose of decorating NPs, PEG seems to
can improve bioavailability of therapeutic agents and
reduce systemic toxicity. Therefore, antitumor efficacy be an ideal candidate and has been extensively used
can be enhanced by multi-functional CSNPs or mul- to coat the surface of NPs, as it successfully weakens
tiple drug loading strategy. For example, co-delivery the uptake by cells of mononuclear phagocytic system
of paclitaxel and survivin shRNA-expressing plasmid (MPS) and leads to prolonged blood circulation time.65
(iSur-pDNA) by folate-modified amphiphilic linoleic acid Some CSNPs also display prolonged blood circulation
and poly(-malic acid) double grafted CSNPs exhib- time after PEG modification. Parveen et al. reported that
ited enhanced antitumor efficacy and prolonged survival PLGA-CS-PEG nanoparticles showed dramatic prolonga-
period as compared with single delivery of PTX or tion in blood circulation, as well as reduced macrophage
iSur-pDNA. 56 uptake, with only a small amount of the nanoparticles
sequestered in the liver.58 Long blood circulation of drug
Improving Special Targeting Tumor Tissue also has great advantage in the treatment of blood malig-
According to the targeting pattern, therapeutic nanopar- nancies. Termsarasab et al. developed doxorubicin (DOX)-
ticles are commonly divided into passive and active loaded nanoparticles based on PEG-conjugated chitosan
targeting nanoparticles. As mentioned above, passive tar- oligosaccharide-arachidic acid (CSOAA-PEG) for poten-
geting nanoparticles accumulation in tumor tissues mainly tial application to leukemia therapy.64 In vivo clearance
depends on the enhanced permeation and retention. Active rate of DOX from the CSOAA-PEG nanoparticle group
targeting based on ligand-receptor interactions is an impor- was slower than other groups, thereby extending the cir-
tant way of increasing specific targeting ability of drug culation period.

1592 J. Biomed. Nanotechnol. 12, 1585–1603, 2016


Fu et al. Chitosan Nanoparticles in Cancer Treatment

Reversal of Multi-Drug Resistance MCF-7/ADR tumors and downregulate P-gp expression to


Multi-drug resistance (MDR) is a major barrier for lim- sensitize cancer cells after intravenous administration.71
iting the therapeutic effects of chemotherapeutic agents
in cancer treatment. Once a patient suffers from MDR, Crossing Blood-Brain Barrier
the therapy efficiency is reduced and leads to the fail- As the biological interface between the brain and the
ure of the treatment. P-glycoprotein-mediated resistance blood, the blood-brain barrier (BBB) takes responsibil-
is the most extensively studied MDR pathway.66 It has ity for transporting essential nutrients and protecting the
been confirmed that nanoparticles may be able to reverse brain.72 The BBB constitutes an efficient organization of
the drug resistance because it may avoid recognition by tight junctions between endothelial cells in the brain tis-
the P-glycoprotein (P-gp) efflux pump by means of being sues. The barrier can prevent harmful substances including
enveloped in an endosome when entering the cell, leading chemotherapeutic agents entering into the brain intersti-
to high intracellular drug concentrations. 67–69 tium and protect the cells of nervous system.73 74 However,
To overcome MDR, Jin et al. used amphiphilic the existence of BBB brings tremendous negative impact
N -octyl-O-sulfate chitosan (NOSC) copolymer to pre- on therapeutic effects of brain tumors when chemother-
pare paclitaxel-encapsulated micelles (PTX-M). Owing to apeutic drugs are used. Therefore, various nanoparti-
the combination of the inhibiting P-gp effect of NOSC cles are being the optional strategies for overcoming the
problem.75–77 Especially, it has been demonstrated that
and the bypassing P-gp action of the intact PTX-M, the
cationic nanoparticles showed prominent efficacy in per-
micelles had superior blood persistence, tumor accumu-
meating the BBB due to their cationic charge.72 78
lation, and therapeutic efficacy after intravenous injection
As a cationic polymer, CSNPs receive much attention
into the tumor-bearing mice (Fig. 4).38 Besides load-
in preparation of nanoformulations or nanoprobes for the
ing of the chemotherapeutic agents, CSNPs are also
treatment and diagnosis of brain cancers. Veiseh et al.
used to deliver genes for the purpose of MDR reversal. used chitosan as a linker and stabilizer to develop a tar-
Zhang et al. used magnetic-Fe3O4 chitosan nanoparticle- geting nanoprobe (NPCP-CTX-Cy5.5) for the purpose of
encapsulated MDR1 siRNA to investigate the effects of selective accumulation in brain tumors across the BBB.72
MDR1 gene on the reversal of MDR in the glioblastoma The nanoprobe is comprised of an iron oxide nanoparti-
cell line BT325. Their results revealed that the expres- cle coated with a PEGylated chitosan-branched copolymer,
sion of MDR1 at both the mRNA Delivered
and P-gp by protein
Ingentalevel
to: Maina CID is 80004805
targeting (JPP)
ligand, chlorotoxin (CTX), and a near-IR flu-
IP: 146.185.205.29
decreased, which increased sensitivity On: Wed,
to chemotherapy 07 Dec 2016 04:41:09
orophore, Cy.5.5. The magnetofluorescent nanoprobe was
in vitro.70 Yhee et al. also confirmedCopyright: American Scientific Publishers
that the thiolated able to traverse the BBB and remain in brain tumors
glycol chitosan could deliver Pgp-targeted poly-siRNA for 70 hours. Stephen et al. developed redox-responsive
and form stable nanoparticles. The resulting nanoparticles magnetic nanoparticles by using chitosan-PEG copolymer
could not only protect siRNA molecules from enzymatic to coat magnetic iron oxide nanoparticles for targeted
degradation, but also accumulate in adriamycin-resistant convection-enhanced delivery (CED) of O6 -benzylguanine

Figure 4. Scheme of collaborative mechanism-induced improvement on the PTX-resistant cellular uptake and therapeutic effi-
cacy of PTX by PTX-M: passive targeting of the intact PTX-M to the tumor tissues by the EPR effect (A) and the mechanism of
enhanced accumulation of PTX inside tumor cells by PTX-M via a combination mechanism of the inhibiting P-gp effect of NOSC
and the bypassing P-gp action of the intact PTX-M (B). Reproduced with permission from [38], X. Jin, et al., Paclitaxel-loaded
N-octyl-O-sulfate chitosan micelles for superior cancer therapeutic efficacy and overcoming drug resistance. Mol. Pharm. 11, 145
(2014). © 2014, American Chemical Society.

J. Biomed. Nanotechnol. 12, 1585–1603, 2016 1593


Chitosan Nanoparticles in Cancer Treatment Fu et al.

to brain tumors.79 The NPs remained within the tumor mesostructured Fe3 O4 /CSNPs by a facile solvothermal
boundary at 48 hours post CED. The in vivo efficacy of the approach, and then used it to load DOX because chi-
NPs in combination with temozolomide showed a 3-fold tosan can act as a host molecule to form the NH2 Zn(II)
increase in survival as compared to untreated animals. DOX coordination architecture.88 To obtain dual imaging
Although the specific mechanisms for BBB passage are of near-infrared (NIR) and magnetic resonance imaging
yet to be elucidated, it has been proven that P-glycoprotein (MRI), Lee et al. encapsulated oleic acid-decorated iron
plays a vital role in the BBB. P-gp is an ATP-dependent oxide nanoparticles (ION) in oleic acid-conjugated chi-
drug transport protein that is expressed in cells at the tosan (oleoyl-chitosan) (Fig. 5).36 During in vivo evalua-
anatomical and physiological barriers in mammalian tis- tion, both NIR and MR imaging showed the detectable
sues and also in malignant cells.80 81 Much effort has signal intensity and enhancement in tumor tissues via
focused on finding ways to overcome drug efflux from enhanced permeability and retention effect after intra-
P-gp expressing cells. Therefore, the strategies to avoid venous injection of ION-loaded Cy5.5-conjugated oleoyl-
P-gp mediated drug efflux at the BBB such as the use of chitosan nanoparticles. Ex vivo fluorescence images and
specific inhibitors, altering the gene regulation and injec- Prussian blue staining images also confirmed that the
tion of siRNA, are being used to reduce the expression of nanoparticles accumulated in tumor tissues. Sun et al.
P-gp in brain endothelial cells.72 Mallmo et al. developed modified the surface of Au nanoparticles with biocompat-
siRNA-CSNPs to silence P-gp in a brain BBB model.82 ible glycol chitosan for tumor-targeting imaging. Several
The nanoparticles led to a significant reduction in P-gp tumor sites of metastatic liver cancer were successfully
substrate efflux and improved the delivery and efficacy of located in the CT images when glycol chitosan-AuNPs
doxorubicin. (0.1 ml, 50 mg Au/ml) were injected intravenously into
the mice.89
Diagnosis, Detection and Imaging Chitosan can also be transformed into a nanoprobe
With the process of nanomedicine and molecular imag- for use in fluorescence imaging. Ryu et al. developed
ing, a variety of tumor-targeting nanoparticles have been a CB-sensitive nanoprobe (CB-CSNP) consisting of self-
designed and extensively studied for cancer theranostics.83 quenched CB-sensitive fluorogenic peptide probes conju-
The functional NPs can chemically interact with biomark- gated onto the surface of tumor-targeting glycol chitosan
ers to alter the signals for imaging and provide biological nanoparticles.90 The prepared CB-CSNP had a spherical
Delivered by Ingenta
information on pathological lesions. Therefore, they areto: Main CID is 80004805
nanoparticle structure(JPP)
with a diameter of 280 nm and its
IP: 146.185.205.29 On: Wed, 07 Dec 2016 04:41:09
fluorescence intensity was strongly quenched in physiolog-
expected to achieve early diagnosis and personalized ther-
Copyright: American Scientific Publishers
apy in cancer treatment in the near future.84 ical condition. Evaluation of CB-CSNP in three metastatic
Owing to excellent biocompatibility and available func- mouse models demonstrated the potential to discriminate
tional groups on chitosan, it is often used to encapsu- metastases in vivo through non-invasive CB molecular
late or decorate other nanoparticles for preparing various imaging.
multifunctional nanoparticles. For the purpose of disease
diagnosis and detection, Yang et al. developed a high- Photodynamic Therapy and Imaging
performance nanoparticle by using alginate to physically Photodynamic therapy (PDT) is becoming a promising
complex with folic acid-modified chitosan for fluorescent and non-invasive method for cancer treatment owing to its
endoscopic detection of colorectal cancer.85 Zhu et al. unique therapeutic characteristics (Fig. 6).91 Under light
developed a probe using nanoparticles of miR-155 MB irradiation at certain wavelength, photosensitizer (PS) pro-
self-assembled with chitosan (CS-miR-155 MB) to image duces cytotoxic singlet oxygen (1 O2  to kill tumor cells
86
the expression of miR-155 in lung cancer cells. The through apoptosis or necrosis.51 92 Moreover, the selec-
CSNPs showed higher fluorescence intensity and trans- tive accumulation of photosensitizers in tumor tissues also
fection efficiency, thus can be used for detecting miRNA can be employed in photodynamic imaging (PDI) because
expression in living cells. Hajdu et al. prepared gadolin- the photosensitizers can provide an intense fluorescence
ium (Gd) ions loaded nanoparticles using poly-gamma- signal.93 94 However, the photosensitizers are greatly lim-
87
glutamic acid (-PGA) and chitosan biopolymers. MRI ited in clinical use owing to their poor water solubility,
measurements and MRI/PET fusion images revealed that non-specific skin phototoxicity, inadequate tumor-targeting
the targeting NPs could accumulate in tumors and provide selectivity, etc. One way for overcoming these shortcom-
a suitable means for the early diagnosis of tumors based ings is the use of nanocarriers to deliver photosensitizers
on their overexpression of folate receptors. for enhancing their tumor specificity by the so-called EPR
Chitosan is commonly used to coat on surfaces of effect.95 Therefore, chitosan is often used as coating poly-
the magnetic iron oxide nanoparticles for the preparation mer to modify photosensitizers because of its nontoxicity,
of various magnetic resonance imaging contrast agents excellent biocompatibility, good water-solubility and avail-
for molecular imaging. Zhao et al. developed a host- ability for further modification of functional groups.51 95
metal-guest coordination-bonding system to co-deliver pH- As a key component of PDT, the most used
responsive drug DOX and Fe3 O4 . They prepared the photosensitizers include protoporphyrin IX (PpIX),

1594 J. Biomed. Nanotechnol. 12, 1585–1603, 2016


Fu et al. Chitosan Nanoparticles in Cancer Treatment

Figure 5. In vivo NIRF images of U87MG-bearing mice. The images were obtained (a) before injection and at (b) 1 h, (c) 3 h,
and (d) 5 h after injection of Ion-Cy5.5-oleyl-chitosan nanoparticles. (e) Ex vivo NIRF images of major tissues excised from the
mice at 5 h postinjection. (f) A quantification of the ex vivo tissues was recorded as average radiance (p/s/cm2/sr). All data are
represented as mean ± SD (n = 3). Reproduced with permission from [36], C. M. Lee, et al., Oleyl-chitosan nanoparticles based
on a dual probe for optical/MR imaging in vivo. Bioconjug. Chem. 22, 186 (2011). © 2011, American Chemical Society.
Delivered by Ingenta to: Main CID is 80004805 (JPP)
IP: 146.185.205.29 On: Wed, 07 Dec 2016 04:41:09
pyropheophorbide a (Ppa), and other porphyrin derivatives.
Copyright: nanoparticles
American Scientific (UCNP-Ppa-RGD).51 The prepared nano-
Publishers
They all show excellent tumor-targeting specificity and structure exhibited low dark toxicity and high phototoxi-
photodynamic imaging ability in many tumor models. Lee city against U87-MG cells upon 980 nm laser irradiation
et al. conjugated the photosensitizer PpIX with glycol chi- at an appropriate dosage. The study of UCNPs has the
tosan polymer to prepare the tumor-targeting nanoparticles potential for cancer theranostics, individualized therapeu-
(PpIX-GC-NPs) for photodynamic imaging and therapy.93 tics, multimodality medicine, etc.
The PpIX-GC-NPs showed the self-quenching effect that
is in the ‘off’ state with no fluorescence signal and photo- Thermotherapy
toxicity with light exposure. After in vivo administration, Because of the thermosensitivity of tumor cells, ther-
the total photon counts of PpIX-GC-NPs in tumor tissue motherapy has been an important support to other
are 1.41 and 2.1 folds higher than that of PpIX-CNP and forms of cancer treatments in clinics. Taking the advan-
free PpIX-treated mice, respectively. tage of passive/active targeting ability of nanoparticles,
However, in order to overcome the limited therapeutic chemotherapeutic agents and functional nanoparticles can
depth of current PDT technologies, upconversion nanopar- be encapsulated simultaneously into CSNPs for combina-
ticles have gained increasing attention in recent years tion therapy. As a kind of thermotherapy, magnetic fluid
due to their unique upconversion luminescent properties.96 hyperthermia (MFH) is a new approach based on nan-
They enable the conversion of low-energy photons (NIR otechnology to deposit heat power in deep tissues by over-
photons) into high-energy photons (visible to UV photons) coming limitations of conventional heat treatments. After
via the multi-photon processes.97 98 Therefore, the specific infiltration into the target tissues with nanosized mag-
characteristics of upconversion nanoparticles including low netic particles, the power of an alternating magnetic field
toxicity, weak background fluorescence, high detection is transformed into heat.101 For the purpose of combi-
sensitivity, and deep light-penetration in tissues endow nation hyperthermia with chemotherapy for the treatment
them with great potential for use in in vivo bioimaging, of liver cancer, Li et al. developed carboplatin-Fe@C-
NIR-triggered drug/gene delivery applications and pho- loaded CSNPs by reverse microemulsion method using the
todynamic therapy.99 100 Zhou et al. used pyropheophor- nanocages Fe@C as magnetic cores and chitosan as the
bide a (Ppa) and RGD peptide c (RGDyK) to modify matrix.102 In vivo experiments showed that tumor tempera-
chitosan for preparation of NIR-to-visible upconversion ture reached 42.6 ± 0.2  C within 10 min in the alternating

J. Biomed. Nanotechnol. 12, 1585–1603, 2016 1595


Chitosan Nanoparticles in Cancer Treatment Fu et al.

Choi et al. developed a very effective hyperthermia system


based on Pluronic F68-conjugated chitosan and GNRs for
photothermal cancer therapy. The prepared nanoplatform
has a reasonable tumor-targeting efficiency and is highly
effective and safe for transducing NIR light into localized
heat.104

Gene Therapy
Gene therapy holds a promising alternative for can-
cer treatment. However, the major shortcoming of gene
therapy is the gene transfection rate.107 Currently, there
are two main types of vectors that are used in gene ther-
apy, viral or non-viral gene delivery systems. Although
the viral gene delivery system shows a high transfec-
tion yield, it still has many disadvantages, such as onco-
genic effects and immunogenicity.108–110 Therefore, as the
alternate, nonviral gene delivery system gains importance.
Figure 6. Schematic diagram of enhanced PDT by utiliz- Especially, numerous synthetic chemicals, natural poly-
ing the surface plasmonic effect of the AuNR to simul- mers, or lipids have been used as nonviral vectors for
taneously increase the absorption coefficient and reduce
enhancing the efficiency of gene delivery.111 Among them,
photo1degradation of the photodynamic dye ICG. Meso-
porous silica provided the second photoprotection for ICG by as a natural nontoxic polysaccharide, chitosan has the
facilitating the formation of ICG aggregates. The double pho- potential for gene delivery applications. Its biodegrad-
toprotection will significantly enhance the stability of loaded- ability, biocompatibility, and the ability to protect DNA
ICG in comparison with the single photoprotection of silica against DNase degradation are added advantages.107
shell. Reprinted with permission from [91], Y. Li, et al., Local- RNAi-based therapy is a highly specific method for
ized electric field of plasmonic nanoplatform enhanced pho-
todynamic tumor therapy. ACS Nano 5, 11529 (2014). © 2014,
gene silencing which holds a dominant position in cancer
American Chemical Society. gene therapy. However, the delivery efficiency of free short
Delivered by Ingenta to: Maininterfering
CID is 80004805
RNA (siRNA)(JPP) is quite low and most of the
IP: 146.185.205.29 On: Wed, 07 Dec 2016 04:41:09
free siRNA is rapidly degraded following i.v. injection.112
magnetic field after injection; and theCopyright:
temperatures American
in the Scientific Publishers
Therefore, in order to improve the delivery efficiency
right hepatic lobes and the rectum were significantly lower
and prolong the existence time, chitosan-based nanopar-
than in the tumor and the constant temperature could last
ticles are often used to load siRNA and form a vari-
up to 30 min. The carboplatin-Fe@C-loaded CSNPs can ety of nanocomplexes because its positive charge allows
specifically target liver cancer tissue by static magnetic transport across cellular membranes and subsequent endo-
field and with the application of alternating magnetic field, cytosis. Han et al. developed Arg-Gly-Asp (RGD) peptide-
effectively raise tumor tissue temperature and facilitate labeled chitosan nanoparticles (RGD-CSNP) as a novel
tumor apoptosis. Therefore, the combination of chemother- tumor targeted delivery system for siRNA.113 In vivo tumor
apy and MFH is likely to be a new safer and efficacious vascular targeting was achieved by using RGD-CSP to
therapy model in cancer treatment. deliver growth-promoting gene PLXDC1-targeted iRNA
Photothermal therapy is an attractive technique for treat- into the  3 integrin-positive tumor endothelial cells in
ing solid tumors in aminimally invasive manner by using the A2780 tumor-bearing mice.
NIR laser light-generated heat to destroy tumor cells Some chitosan derivatives also show good DNA-
(Fig. 7).103–105 Light-absorbing materials e.g., photothermal binding capacity and gene therapy efficacy after func-
conducting agents, as the source of photothermal effect, tional modification. For example, Hu et al. prepared
play a key role in photoactivated cancer therapy. Among a non-viral vesicle vector which was composed of
reported NIR absorbents, gold nanocrystals, including gold low-molecular weight polyethylenimine-conjugated stearic
nanorods, silica-cored nanoshells, and gold nanocages, acid-g-chitosan oligosaccharide (CSOSA-g-PEI) micelles.
were most extensively investigated due to their excellent The micelles had much lower cytotoxicity and corre-
biocompatibility and tunable surface plasmon resonance sponding transfection efficiency in comparison with Lipo-
property to convert NIR light into local heat. Duan et al. fectamine 2,000 in both human cancer cells (Hela and
used DOX-conjugated chitosan derivatives to cover gold MCF-7). Moreover, after intravenous injection in tumor-
nanorods (GNRs) for preparation of functional nanocar- bearing mice, CSOSA-g-PEI/plasmid pigment epithelium-
riers (DOX-CS-GNR).106 Because of the combination of derived factor formulation could effectively localize in the
chemical and photothermal effects, the prepared nanocar- tumor tissue and suppress the tumor growth (above 60%
riers showed good optical properties and enhanced antitu- tumor inhibition) without systematic toxicity.114 In addi-
mor activity compared with photothermal therapy alone. tion to those mentioned above, other chitosan-based

1596 J. Biomed. Nanotechnol. 12, 1585–1603, 2016


Fu et al. Chitosan Nanoparticles in Cancer Treatment

Figure 7. (A) Infrared thermal imaging of P-GNRs-DOX (1.0 mg mL−1 , contained 28 g Au mL−1 ) under 2.5 W cm−2 irradiation by
808 nm laser at different time and (B) infrared thermal imaging of tumor under the photothermal heating by 808 nm laser irradiation
in normal saline and P-GNRs-DOX injected mice under 2.5 W cm−2 irradiation. Reprinted with permission from [105], J. F. Liao,
et al., Combined cancer photothermal-chemotherapy based on doxorubicin/gold nanorod-loaded polymersomes. Theranostics
5, 345 (2015). © 2015, Theranostics Publishing Team.

nanoparticles have being widely studied to deliver siRNA an important role in cancer treatment. For example,
for gene silencing and these nanocomplexes have shown lactosyl-norcantharidin TMC nanoparticles (Lac-NCTD-
great potential in gene therapy.115–120 TMC-NPs) were prepared using an ionic cross-linkage
In addition, the use of chitosan vectors to deliver process.130 The Lac-NCTD-TMC-NPs had an average
short hairpin RNA (shRNAs) to knock-down genes particle size of about 120.6 nm and it showed the
has attracted considerable attention in cancer treat- strongest antitumor activity on the murine hepatocar-
ments. Chen et al. designed a novel nanoparticle cinoma 22 subcutaneous models. Hu et al. prepared
(Au-PEI/CS-Aco/PEI/shRNA) for delivering shRNA by chondroitin sulfate-chitosan (ChS-CS) nanoparticles and
Delivered by Ingenta to: Main CID is 80004805 (JPP)
using a three-layered polyethyleneimine (PEI)-coated gold
IP: 146.185.205.29 On: Wed, investigated
07 Dec 2016their 04:41:09
cellular uptake, cytotoxicity, and transep-
nanocomplex interlaid with a pH-responsive charge- Scientific
Copyright: American ithelial transport in human epithelial colorectal adenocar-
Publishers
reversible chitosan-aconitic anhydride (CS-Aco) as a car- cinoma (Caco-2) fibroblasts.131
rier. The prepared nanocomplexes displayed higher shRNA In addition to natural extracts, many first-line
delivery efficiency than 25 kDa PEI, and efficiently deliv- chemotherapeutic agents are also encapsulated in CSNPs
ered shABCG2 to tumors and markedly silenced the for cancer treatment owing to good biodegradability and
expression of ABCG2.121 biocompatibility of chitosan. Among them, 5-FU42 44 57 132
and cisplatin133 134 are well studied. For example, Xu et al.
CHITOSAN NANOPARTICLES IN developed an effective pH and oxidation dual-responsive
DRUG DELIVERY carrier by using ferrocene-modified chitosan oligosac-
Encapsulation of Chemotherapeutic charide nanoparticles for 5-FU delivery.132 Kim et al.
Agents Into Chitosan used hydrophobic cholanic acid-modified glycol chitosan
Chemotherapeutic agents play a central role in can- (HGCs) to encapsulate cisplatin (CDDP) and they believed
cer intervention. Because of their significant antitumor that HGC nanoparticles are a promising carrier for the
effect, various chemotherapeutic drugs, including natural anticancer drug CDDP.133 Arya et al. prepared herceptin-
or synthetic agents, were intensively investigated. As a conjugated gemcitabine-loaded CSNPs to target the deliv-
typical example of chemotherapeutic agent of plant ori- ery of gemcitabine for the treatment of pancreatic cancer.49
gin, paclitaxel was encapsulated into CSNPs by many
researchers. These systems showed a good controlled Directly Conjugated Chemotherapeutic
release behavior and enhanced antitumor efficacy in many Drugs with Chitosan Backbone
tumor cells such as MDA-MB231 human breast cancer Even though chitosan is extensively used as a drug car-
cells, B16F10, A2780 cells, Caco-2 cells, HepG2 cells, rier for delivery of various chemotherapeutic drugs, it only
etc.54 122–125 Additionally, other plant-derived anticancer acts as a vehicle to load drugs and not play a role in can-
agents and their synthetic derivatives, such as baicalein, cer treatment. Therefore, a prodrug strategy was proposed
camptothecine, curcumin, and resveratrol, were loaded on owing to rich reactive groups on chitosan backbone. Some
chitosan nanocarriers for tumor therapy.126–129 the hydrophobic anticancer drug can be directly conju-
Anticancer compounds of animal origin have also gated onto hydrophilic chitosan chains resulting in better
shown excellent antitumor activity and could play hydrophilicity and bioavailability of drugs. For example,

J. Biomed. Nanotechnol. 12, 1585–1603, 2016 1597


Chitosan Nanoparticles in Cancer Treatment Fu et al.

DOX was simply conjugated with chitosan via an acid-


cleavable hydrazone bond.135 The prepared prodrugs could
directly disperse in an aqueous solution without the use
of any organic solvent. The resulting Chitosan-hz-DOX
nanoparticles with size lower than 200 nm had an excel-
lent pH-sensitive drug release property as well as antitu-
mor activity. The Chitosan-hz-DOX particles were stable
at a neutral pH while disassembled at acidic pHs, result-
ing in a fast release of DOX. This was desired for the
internalization process of prodrug nanoparticles into can-
cer cells. Lu et al. developed a series of melphalan-O-
carboxymethyl chitosan conjugates. These conjugates had
a remarkably improved water solubility (160–217 times
that of melphalan solubility) and 52–70% of cytotoxicity Figure 8. Schematic illustration of induced apoptosis in HT
against RPMI8226 cells in vitro as compared with free 29 cells by chitosan nanocarriers loading silver nanoparti-
melphalan.136 cles. Reprinted with permission from [137], P. Sanpui, et al.,
Induction of apoptosis in cancer cells at low silver nanopar-
ticle concentrations using chitosan nanocarrier. ACS Appl.
Encapsulation of Metal Compounds Mater. Interf. 3, 218 (2011). © 2011, American Chemical
Several metals and their compounds find special appli- Society.
cations in cancer diagnosis and treatment owing to their
unique properties. For example, platinum compounds have
significant antitumor efficacy and broad spectrum anti- the therapeutic efficacy and biocompatibility are greatly
cancer activity attributable to their remarkable cytotoxic- weakened due to their inherent instability, poor pharma-
ity against tumor cells. Gold nanoparticles are often used cokinetics, and potential toxicity.139–141 Therapeutic pro-
in NIR photothermal therapy due to their tunable sur- teins have high molecular weight (MW > 1,000 Da) and
face plasmon resonance property to convert NIR light into good hydrophilicity which significantly affect their ability
local heat.106 Magnetic iron oxide nanoparticles
Delivered canto:
by Ingenta beMaintoCID
cross
is cell membranes.
80004805
142
(JPP) Moreover, proteins are easily
used as a magnetic resonance imaging agent and tumor-
IP: 146.185.205.29 On: Wed, and
07rapidly
Dec 2016 removed from the bloodstream after under-
04:41:09
Copyright:
targeting drug carriers because they have American
excellent mag- Scientific Publishers
going proteolytic cleavage in vivo.143 Additionally, the
netic response.23 70 conditions of production and storage of protein drug for-
In addition to above mentioned metallic compounds, mulations may cause adsorption, aggregation, and denatu-
several other metal nanoparticles and compounds are also ration. This reduces their effectiveness considerably under
encapsulated in chitosan for cancer therapy. Several cop- in vivo conditions.144 145
per compounds have demonstrated anticancer activity. To circumvent these issues and further improve the
For example, the complex containing 1,3-diketone group bioavailability of proteins, a large number of investigations
of copper ion and acetylacetone (CuAA) was delivered have been carried out by employing numerous methods,
by folic acid-tagged hydrophobic-modified CSNPs.35 The including addition of nonionic surfactants, metal chelators,
NPs could induce apoptosis and reduce mitochondrial enzyme inhibitors, etc.142 Furthermore, many chitosan-
membrane potential of cancer cells. It could also be suc- based nanoparticles have been designed and extensively
cessfully delivered to folate receptor over-expressed cancer tailored to deliver various proteins according to the types
cells. Sanpui et al. used chitosan nanocarriers (CS NCs) of proteins and their practical purposes.146 147 In Caco-2
to deliver silver nanoparticles (Ag NPs) for induction of cell and rat model, N -trimethyl chitosan chloride (TMC)
apoptosis in human colon cancer cells (HT 29) (Fig. 8).137 was confirmed as a potent mucosal permeation enhancer
At a much lower concentration (0.33 g/mL) of Ag NPs, for the peptide drug buserelin at neutral pH. Coadminis-
the viability of HT 29 cells was reduced to 50%. The tration of buserelin with TMCs resulted in mean bioavail-
oxidative stress and mitochondrial dysfunction were found ability values between 6 and 13% which was greatly
to play an important role in the apoptosis induced by the higher than that of intraduodenally applied buserelin with
Ag-CS NCs. 0.8% absolute bioavailability.148 Self-assembled glycol
chitosan nanoparticles also show high efficacy for sus-
Encapsulation of Protein tained and prolonged delivery of peptide drugs in cancer
Therapeutic peptides/proteins and protein-based antigens therapy.149–151 Kime et al. developed hydrophobically-
are being extensively utilized for the management and cure modified glycol chitosan (HGC) nanoparticles conjugated
of many diseases, particularly oncologic and metabolic with interleukin-4 receptor (IL-4R) binding peptides (I4R),
diseases.138 Although they are frequently used in clinic and and then tested them in mice bearing IL-4R-positive
almost exclusively administered by parenteral injections, tumors. Their study showed that HGC-I4R nanoparticles

1598 J. Biomed. Nanotechnol. 12, 1585–1603, 2016


Fu et al. Chitosan Nanoparticles in Cancer Treatment

exhibited enhanced IL-4R-dependent cellular uptake in Declaration of Interest Statement


tumors compared to nonconjugated nanoparticles, con- The authors declare no conflicts of interest. The authors
sequently leading to better therapeutic and imaging alone are responsible for the content and writing of the
efficacy.151 paper.

Acknowledgments: This work was financially sup-


PROBLEMS AND OUTLOOK ported by the Union Project of Luzhou City and Luzhou
Even though chitosan has many advantages in drug deliv- Medical College (14JC0144, 2013LZLY-J40), Young
ery, there still are several problems which can not be Scholars Fund of Luzhou Medical College (2012QN-14),
ignored. One problem is the potential toxicity caused by Science and Technology Project of Luzhou City (2013-
modification of chitosan. Chitosan has poor solubility and 134), Youth Talent Fund of the Affiliated Hospital of
unmodified CSNPs can encapsulate only some hydrophilic Luzhou Medical College (2013-60).
drugs. In order to make chitosan encapsulate hydrophobic
drugs easily, structure modification is necessary. There-
REFERENCES
fore, various modifiers, catalysts or crosslinking agents
18 1. B. Wilson, T. V. Ambika, K. P. R. Dharmesh, J. L. Jenita, and
are widely used to decorate CSNPs. Moreover, some S. R. Priyadarshini, Nanoparticles based on albumin: Preparation,
toxic organic solvents are also used during the process of characterization and the use for 5-flurouracil delivery. Int. J. Biol.
modification. The use of these chemicals may bring obvi- Macromol. 51, 874 (2012).
ous side-effects due to their intrinsic toxicity. Therefore, 2. S. Cecco, M. Aliberti, P. Baldo, E. Giacomin, and R. Leone, Safety
and efficacy evaluation of albumin-bound paclitaxel. Expert Opin.
after each chemical reaction, the unreacted reagents and Drug Saf. 13, 511 (2014).
reactants must be removed completely to prevent confus- 3. S. Nazir, T. Hussain, A. Ayub, U. Rashid, and A. J. MacRobert,
ing results.10 Also, the further investigation is required on Nanomaterials in combating cancer: Therapeutic applications and
the biocompatibility of modified chitosan and its deriva- developments. Nanomedicine 10, 19 (2014).
4. M. P. Patel, R. R. Patel, and J. K. Patel, Chitosan mediated targeted
tives. Another problem is retaining the encapsulation effi- drug delivery system: A review. J. Pharm. Pharm. Sci. 13, 536
ciency through mass production of drug loading systems. (2010).
Commonly, high drug loading efficacy and encapsulation 5. F. R. Wurm and C. K. Weiss, Nanoparticles from renewable poly-
efficiency can be achieved at a small scale in lab. How- mers. Front Chem. 2, 49 (2014).
6. D. isAgudelo,
Delivered by Ingenta to: Main CID 80004805 (JPP)
S. Sanyakamdhorn, S. Nafisi, and H. A. Tajmir-
ever, it is very difficult to maintain the same high load-
IP: 146.185.205.29 On: Wed, 07Riahi,
DecTransporting
2016 04:41:09antitumor drug tamoxifen and its metabolities,
ing efficacy and stability during a large-scale production.
Copyright: American Scientific Publishers and endoxifen by chitosan nanoparticles. PLoS
4-hydroxytamoxifen
Additionally, although nanosized particles have a wide ONE 8, e60250 (2013).
range of potential biomedical and pharmaceutical applica- 7. A. Rampino, M. Borgogna, P. Blasi, B. Bellich, and A. Cesaro,
Chitosan nanopraticles: Preparation, size evolution and stability. Int.
tions, the unique physicochemical properties of nanopar- J. Pharm. 455, 219 (2013).
ticles may cause potential risks to human body and the 8. K. Bowan and K. W. Leong, Chitosan nanoparticles for oral drug
environment. Some studies have demonstrated that CSNPs and gene delivery. Int. J. Nanomed. 1, 117 (2006).
may also exhibit different cytotoxicity profiles depending 9. V. Dodane, M. A. Khan, and J. R. Merwin, Effect of chitosan on
epithelial permeability and structure. Int. J. Pharm. 182, 21 (1999).
on the properties of the parent material used to synthe-
10. M. Dash, F. Chiellini, R. M. Ottenbrite, and E. Chiellinni, Chitosan-
size the nanoparticles, the size of the nanoparticles and A versatile semi-synthetic in biomedical applications. Prog. Poly-
the interacting cell types.10 15 Therefore, potential toxicity mer Sci. 36, 981 (2011).
of chitosan-based nanoformulations should be taken into 11. X. Li, S. Chen, B. Zhang, M. Li, K. Diao, Z. Zhang, J. Li, Y. Xu,
X. Wang, and H. Chen, In situ injectable nano-composite hydrogel
consideration including their in vivo distribution in normal
composed of curcumin, N ,O-carboxymethyl chitosan and oxidized
tissues and organs. alginate for wound healing application. Int. J. Pharm. 437, 110
Even though a large-scale application of CSNPs in (2012).
clinic may take some time, the advances in nanotech- 12. S. C. C. C. Miranda, G. A. B. Silva, R. C. R. Hell, M. D. Martins,
nology and polymer chemistry is poised to unravel the J. B. Alves, and A. M. Goes, Three-dimensional culture of rat
BMMSCs in a porous chitosan-gelatin scaffold: A promising asso-
full potential of the chitosan-based nanodrug delivery ciation for bone tissue engineering in oral reconstruction. Arch.
systems have the great potential for the development Oral Biol. 56, 1 (2011).
of multifunctional “smart” nanoparticles that may facil- 13. L. Casettari and L. Illum, Chitosan in nasal delivery systems for
itate the realization of individualized cancer therapy.18 therapeutic drugs. J. Control. Release 190, 189 (2014).
14. Y. L. Hu, W. Qi, F. Han, J. Z. Shao, and J. Q. Gao, Toxicity eval-
So far, a variety of multifunctional CSNPs have been uation of biodegradable chitosan nanoparticles using a zebrafish
evaluated for application to cancers detection, diagnosis, embryo model. Int. J. Nanomed. 6, 3351 (2011).
simultaneous treatment and in vivo imaging. Although 15. J. W. Loh, G. Yeoh, M. Saunders, and L. Y. Lim, Uptake and
some barriers still exist in the development of chitosan- cytotoxicity of chitosan nanoparticles in human liver cells. Toxicol.
Appl. Pharm. 249, 148 (2010).
based nanoformulations, we still believe that chitosan 16. M. Huang, E. Khor, and L. Y. Lim, Uptake and cytotoxicity of
nanocarriers have broad application prospects in cancer chitosan molecules and nanoparticles: Effects of molecular weight
therapy. and degree of deacetylation. Pharm. Res. 21, 344 (2004).

J. Biomed. Nanotechnol. 12, 1585–1603, 2016 1599


Chitosan Nanoparticles in Cancer Treatment Fu et al.

17. L. Qi, Z. Xu, and M. Chen, In vitro and in vivo suppression of 35. A. Pramanik, D. Laha, P. Pramanik, and P. Karmakar, A novel
hepatocellular carcinoma growth by chitosan nanoparticles. Eur. drug “copper acetylacetonate” loaded in folic acid-tagged chitosan
J. Cancer 43, 184 (2007). nanoparticle for efficient cancer cell targeting. J. Drug Target.
18. T. Sonia and C. Sharma, Chitosan and its derivatives for drug deliv- 22, 23 (2014).
ery perspective. Adv. Polym. Sci. 243, 23 (2011). 36. C. M. Lee, D. Jang, J. Kim, S. J. Cheong, E. M. Kim, M. H. Jeong,
19. A. Bianco, K. Kostarelos, and M. Prato, Applications of carbon S. H. Kim, D. W. Kim, S. T. Lim, M. H. Sohn, Y. Y. Jeong, and
nanotubes in drug delivery. Curr. Opin. Chem. Biol. 9, 674 (2005). H. J. Jeong, Oleyl-chitosan nanoparticles based on a dual probe for
20. C. Li, K. Yang, Y. Zhang, H. Tang, F. Yan, L. Tan, Q. Xie, optical/MR imaging in vivo. Bioconjug. Chem. 22, 186 (2011).
and S. Yao, Highly biocompatible multi-walled carbon nanotube- 37. S. J. Yang, C. F. Lin, M. L. Kuo, and C. T. Tan, Photodynamic
chitosan nanoparticle hybrids as protein carriers. Acta Biomater. detection of oral cancers with high-performance chitosan-based
7, 3070 (2011). nanoparticles. Biomacromolecules 14, 3183 (2013).
21. X. Liu, Y. Zhang, D. Ma, H. Tang, L. Tan, Q. Xie, and S. Yao, 38. X. Jin, R. Mo, Y. Ding, W. Zheng, and C. Zhang, Paclitaxel-loaded
Biocompatible multi-walled carbon nanotube-chitosan-folic acid N -octyl-O-sulfate chitosan micelles for superior cancer therapeu-
nanoparticle hybrids as GFP gene delivery materials. Colloi. Surf. tic efficacy and overcoming drug resistance. Mol. Pharm. 11, 145
B 111, 224 (2013). (2014).
22. M. Gou, K. Men, J. Zhang, Y. Li, J. Song, S. Luo, H. Shi, Y. Wen, 39. N. Bhattarai, H. R. Ramay, S. H. Chou, and M. Zhang, Chitosan
G. Guo, M. Huang, X. Zhao, Z. Qian, and Y. Wei, Efficient inhi- and lactic acid-grafted chitosan nanoparticles as carriers for pro-
bition of C-26 colon carcinoma by VSVMP gene delivered by longed drug delivery. Int. J. Nanomed. 1, 181 (2006).
biodegradable cationic nanogel derived from polyethyleneimine. 40. S. J. Mao, Y. Q. Bi, H. Jin, D. P. Wei, R. He, and S. X. Hou,
Acs Nano 4, 5573 (2010). Preparation, characterization and uptake by primary cultured rat
23. S. H. Yuk, K. S. Oh, S. H. Cho, B. S. Lee, S. Y. Kim, B. K. Kwak, hepatocytes of liposomes surface-modified with glycyrrhetinic acid.
K. Kim, and I. C. Kwon, Glycol chitosan/heparin immobilized iron Pharmazie 62, 614 (2007).
oxide nanoparticles with a tumor-targeting characteristic for mag- 41. Q. Tian, X. H. Wang, W. Wang, C. N. Zhang, P. Wang, and Z. Yuan,
netic resonance imaging. Biomacromolecules 12, 2335 (2011). Self-assembly and liver targeting of sulfated chitosan nanoparti-
24. C. K. Lai, Y. L. Lu, J. T. Hsieh, S. C. Tsai, C. L. Feng, Y. S. cles functionalized with glycyrrhetinic acid. Nanomedicine 8, 870
Tsai, P. C. Tsai, H. L. Su, Y. H. Lin, and C. H. Lai, Development (2012).
of chitosan/heparin nanoparticle-encapsulated cytolethal distending 42. M. Cheng, H. Chen, Y. Wang, H. Xu, B. He, J. Han, and
toxin for gastric cancer therapy. Nanomedicine 9, 803 (2013).
Z. Zhang, Optimized synthesis of glycyrrhetinic acid-modified chi-
25. L. Yang, S. Gao, S. Asghar, G. Liu, S. Jong, X. Wang, Q. Ping,
tosan 5-fluorouracil nanoparticles and their characteristics. Int. J.
C. Zhang, and Y. Xiao, Hyaluronic acid/chitosan nanoparticles for
Nanomed. 9, 695 (2014).
delivery of curcuminoid and its in vitro evaluation in glioma cells.
43. X. P. He, B. W. Zhu, Y. Zang, J. Li, G. R. Chen, H. Tian, and
Int. J. Biol. Maromol. 72, 1391 (2015).
Y. T. Long, Dynamic tracking of pathogenic receptor expression
26. X. Deng, M. Gao, J. Zhang, K.Delivered
Hu, Z. Yin, by Ingenta to: Main CIDof is
live80004805 (JPP)glycoanthraquinone-decorated graphene
Z. Zhou, X. Xiao,
cells using pyrenyl
Y. Yang, W. Sheng, Y. Wu, and Y.IP: 146.185.205.29 On: Wed, 07electrodes.
Zeng, Hyaluronic acid-chitosan Dec 2016 04:41:09
Chem. Sci. 6, 1996 (2015).
nanoparticles for co-delivery of MiR-34aandCopyright:
doxorubicin American
in therapy Scientific Publishers
44. M. Cheng, B. He, T. Wan, W. Zhu, J. Han, B. Zha, H. Chen,
against triple negative breast cancer. Biomaterials 35, 4333 (2014).
F. Yang, Q. Li, W. Wang, H. Xu, and T. Ye, 5-Fluorouracil nanopar-
27. M. Fuente, B. Seijo, and M. J. Alonso, Novel hyaluronic acid-
ticles inhibit hepatocellular carcinoma via activation of the p53
chitosan nanoparticles for ocular gene therapy. IOVS 5, 2016
pathway in the orthotopic transplant mouse model. PLoS ONE
(2008).
7, e47115 (2012).
28. Y. Luo and Q. Wang, Recent development of chitosan-based poly-
electrolyte complexes with natural polysaccharides for drug deliv- 45. Q. Wang, L. Zhang, W. Hu, Z. H. Hu, Y. Y. Bei, J. Y. Xu,
ery. Int. J. Biol. Macromol. 64, 353 (2014). W. J. Wang, X. N. Zhang, and Q. Zhang, Norcantharidin-associated
29. D. T. Martin, J. M. Steinbach, J. Liu, S. Shimizu, H. Z. galactosylated chitosan nanoparticles for hepatocyte-targeted deliv-
Kaimakliotis, M. A. Wheeler, A. B. Hittelman, W. M. Saltz- ery. Nanomedicine 6, 371 (2010).
man, and R. M. Weiss, Sruface-modified nanoparticles enhance 46. W. J. Lin and W. Y. Hsu, Pegylation effect of chitosan based poly-
transurothelial penetration and delivery of surviving siRNA in treat- plex on DNA transfection. Carbohyd. Polym. 120, 7 (2015).
ing bladder cancer. Mol. Cancer Ther. 13, 71 (2014). 47. J. M. Yu, W. D. Li, L. Lu, X. Y. Zhou, D. Y. Wang, H. M. Li,
30. T. Zhang, G. Li, L. Guo, and H. Chen, Synthesis of thermo- X. Y. Xu, and J. Chen, Preparation and characterization of galacto-
sensitive CS-g-PNIPAM/CMC complex nanoparticles for controlled sylated glycol chitosan micelles and its potential use for hepatoma-
release of 5-FU. Int. J. Biol. Marcomol. 51, 1109 (2012). targeting delivery of doxorubicin. J. Mater. Sci. 25, 691 (2014).
31. X. Zhang, J. Zhao, Y. Wen, C. Zhu, J. Yang, and F. Yao, 48. R. Villa, B. Cerroni, L. Vigano, S. Margheritelli, G. Abolafio,
Carboxymethyl chitosan-poly(amidoamine) dendrimer core–shell L. Oddo, G. Paradossi, and N. Zaffaroni, Targeted doxorubicin
nanoparticles for intracellular lysozyme delivery. Carbohydr. delivery by chitosan-galactosylated modified polymer microbubbles
Polym. 98, 1326 (2013). to hepatocarcinoma cells. Colllid. Surf. B 110, 434 (2013).
32. Y. Jeong, S. H. Kim, T. Y. Jung, I. Y. Kim, S. S. Kang, Y. H. Jin, 49. G. Arya, M. Vandana, S. Acharya, and S. K. Sahoo,
H. H. Ryu, H. S. Sun, S. Jin, K. K. Kim, K. Y. Ahn, and S. Jung, Enhanced antiproliferative activity of Herceptin (HER2)-conjugated
Polyion complex micelles composed of all-trans retinoic acid and gemcitabine-loaded chitosan nanoparticle in pancreatic cancer ther-
poly(ethylene glycol)-grafted-chitosan. J. Pharm. Sci. 95, 2348 apy. Nanomedicine 7, 859 (2011).
(2006). 50. M. H. Yang, T. W. Chung, Y. S. Lu, Y. L. Chen, W. C. Tsai, S. B.
33. C. Chen, G. Cai, H. Zhang, H. Jiang, and L. Wang, Chitosan- Jong, S. S. Yuan, P. C. Liao, P. C. Lin, and Y. C. Tyan, Activation of
poly(-caprolactone)–poly(ethylene glycol) graft copolymers: Syn- the ubiquitin proteasome pathway by silk fibroin modified chitosan
thesis, self-assembly, and drug release behavior. J. Biomed. Mater. nanoparticles in hepatic cancer cells. Int. J. Mol. Sci. 16, 1657
Res. A 96, 116 (2011). (2015).
34. D. Jeevitha and K. Amarnath, Chitosan/PLA nanoparticles as a 51. A. Zhou, Y. Wei, B. Wu, Q. Chen, and D. Xing, Pyropheophor-
novel carrier for the delivery of anthraquinone: Synthesis, char- bide A and c(RGDyk) comodifed chitosan-wrapped upconversion
acterization and in vitro cytotoxicity evaluation. Colloi. Surf. B nanoparticle for targaeted near-infrared photodynamic therapy. Mol.
101, 126 (2013). Pharm. 9, 1580 (2012).

1600 J. Biomed. Nanotechnol. 12, 1585–1603, 2016


Fu et al. Chitosan Nanoparticles in Cancer Treatment

52. J. Liu, L. Zhang, C. Wang, H. Xu, and X. Zhao, Preparation and 70. P. Zhao, H. Wang, H. Gao, C. Li, and Y. Zhang, Reversal of
characterization of lectin-conjugated chitosan fluorescent nanopar- multidrug resistance by magnetic chitosan-Fe(3)O(4) nanoparticle-
ticles. Mol. Biosyst. 6, 954 (2010). encapsulated MDR(1) siRNA in glioblastoma cell line. Neurol. Res.
53. J. J. Wang, Z. W. Zeng, R. Z. Xiao, T. Xie, G. L. Zhou, X. R. 35, 821 (2013).
Zhan, and S. L. Wang, Recent advances of chitosan nanoparticles 71. J. Y. Yhee, S. Song, S. J. Lee, S. G. Park, K. S. Kim, M. G.
as drug carriers. Int. J. Nanomed. 6, 765 (2011). Kim, S. Son, H. Koo, I. C. Kwon, J. H. Jeong, S. Y. Jeong, S. H.
54. H. Koo, K. H. Min, S. C. Lee, J. H. Park, K. Park, S. Y. Jeong, Kim, and K. Kim, Cancer-targeted MDR-1 siRNA delivery using
K. Choi, I. C. Kwon, and K. Kim, Enhanced drug-loading and self-cross-linked glycol chitosan nanoparticles to overcome drug
therapeutic efficacy of hydrotropic oligomer-conjugated glycol chi- resistance. J. Control. Release. 198, 1 (2015).
tosan nanoparticles for tumor-targeted paclitaxel delivery. J. Con- 72. O. Veiseh, C. Sun, C. Fang, N. Bhattarai, J. Gunn, F. Kievit, K. Du,
trol. Release. 172, 823 (2013). B. Pullar, D. Lee, R. G. Ellenbogen, J. Olson, and M. Zhang,
55. B. Teong, C. Y. Lin, S. J. Chang, G. C. Niu, C. H. Yao, I. F. Chen, Specific targeting of brain tumors with an optical/magnetic reso-
and S. M. Kuo, Enhanced anti-cancer activity by curcumin-loaded nance imaging nanoprobe across the blood-brain barrier. Cancer
hydrogel nanoparticle derived aggregates on A549 lung adenocar- Res. 69, 6200 (2009).
cinoma cells. J. Mater. Sci. Mater. Med. 26, 1 (2015). 73. J. Kreuter, Nanoparticulate systems for brain delivery of drugs. Adv.
56. B. Yu, C. Tang, and C. Yin, Enhanced antitumor efficacy of Drug Deliv. Rev. 47, 65 (2001).
folate modified amphiphilic nanoparticles through co-delivery of 74. M. W. Smith and M. Gumbleton, Endocytosis at the blood-
chemotherapeutic drugs and genes. Biomaterials 35, 6369 (2014). brain barrier: from basic understanding to drug delivery strategies.
57. Z. Ghasemi, R. Dinarvand, F. Mottaghitalab, M. Esfandyari- J. Drug. Target. 14, 191 (2006).
Manesh, E. Sayari, and F. Atyabi, Aptamer decorated hyaluro- 75. C. Li, S. Li, T. Tu, X. Qi, Y. Xiong, S. Du, Y. Shen, J. Tu, and
nan/chitosan nanoparticles for targeted delivery of 5-fluorouracil C. Sun, Paclitaxel-loaded cholesterol-conjugated polyoxyethylene
to MUC1 overexpressing adenocarcinomas. Carbohydr. Polym. sorbitol oleate polymeric micelles for glioblastoma therapy across
121, 190 (2015). the blood-brain barrier. Polym. Chem. 6, 2740 (2015).
58. S. Parveen and S. K. Sahoo, Long circulating chitosan/PEG blended 76. I. P. Kaur, R. Bhandari, S. Bhandari, and V. Kakkar, Potential
PLGA nanoparticle for tumor drug delivery. Eur. J. Pharmacol. of solid lipid nanoparticles in brain targeting. J. Control. Release
670, 372 (2011). 127, 97 (2008).
59. D. E. O. Iii and N. A. Peppas, Opsonization, biodistribution, 77. R. Alyautdin, L. Khalin, M. I. Nafeeza, M. H. Haron, and
and pharmacokinetics of polymeric nanoparticles. Int. J. Pharm.
D. Kuznetsov, Nanoscale drug delivery systems and the blood-brain
307, 93 (2006).
barrier. Int. J. Nanomed. 9, 795 (2014).
60. F. Alexis, E. Pridgen, L. K. Molnar, and O. C. Farokhzad, Factors
78. W. Lu, J. Wan, Z. She, and X. Jiang, Brain delivery property and
affecting the clearance andbiodistribution of polymeric nanoparti-
accelerated blood clearance of cationic albumin conjugated pegy-
cles. Mol. Pharmacol. 5, 505 (2008).
lated nanoparticles. J. Control. Release 118, 38 (2007).
61. M. M. Amiji, Synthesis of anionicDelivered
poly(ethyleneby Ingenta to: Main79.
CIDZ. is
R. 80004805
Stephen, F. M.(JPP)
glycol) derivative
Kievit, O. Veiseh, P. A. Chiarelli, C. Fang,
for chitosansurface modification inIP: 146.185.205.29 On: Wed, 07K.Dec 2016S. J.04:41:09
blood-contacting applications.
Wang, Hatzinger, R. G. Ellenbogen, J. R. Silber, and
Carbohydr. Polym. 32, 193 (1997). Copyright: American Scientific Publishers
M. Zhang, Redox-responsive magnetic nanoparticles for targeted
62. Y. Sheng, C. Liu, Y. Yuan, X. Tao, F. Yang, X. Shan, H. Zhou,
convection-ehanced delivery of O6 -benzylguanine to brain tumors.
and F. Xu, Long-circulatin polymeric nanoparticles bearing a com-
ACS Nano 8, 10383 (2014).
binatorial coating of PEG and water-soluble chitosan. Biomaterials
80. A. H. Schinkel, P-glycoprotein, a gatekeeper in the blood-brain
30, 2340 (2009).
barrier. Adv. Drug Del. Rev. 36, 179 (1999).
63. R. Gref, ckM. Lu, P. Quellec, M. Marchand, E. Dellacherie,
S. Harnisch, T. Blunk, and R. H. Muller, ‘Stealth’ corona-core 81. A. H. Schinkel, J. J. M Smit, O. Tellingen, J. H. Beijnen,
nanoparticles surface modified by polyethylene glycol (PEG): Influ- E. Wagenaar, L. Deemter, C. A. A. M. Mol, M. A. Van der Valk,
ences of the corona (PEG chain length and surface density) and E. C. R. Robanus-Maandag, H. P. J. Riele, A. J. M. Berns, and
of the core composition on phagocytic uptake and plasma protein P. Borst, Disruption of the mouse mdr1a P-glycoprotein gene leads
adsorption. Colloid. Surf. B 18, 301 (2000). to a deficiency in the blood-brain barrier and to increased sensitivity
64. U. Termsarasab, I. S. Yoon, J. H. Park, H. T. Moon, H. J. Cho, and to drugs. Cell 77, 491 (1994).
D. D. Kim, Polyethylene glycol-modified arachidyl chitosan-based 82. J. Mallmo, A. Sandvig, K. M. Varum, and S. P. Strand, Nanopar-
nanoparticles for prolonged blood circulation of doxorubicin. Int. ticle mediated P-glycoprotein silencing for improved drug delivery
J. Pharm. 464, 127 (2014). across the blood-brain barrier: A siRNA-chitosan approach. PLoS
65. G. Fontana, M. Licciardi, S. Mansueto, D. Schillaci, and ONE 8, e54182 (2013).
G. Giammona, Amoxicillin-loaded polyethylcyanoacrylate 83. K. Chen and X. Chen, Design and development of molecular imag-
nanoparticles: Influence of PEG coating on the particle size, drug ing probes. Curr. Top Med. Chem. 10, 1227 (2010).
release rate and phagocytic uptake. Biomaterials 22, 2857 (2001). 84. J. Y. Yhee, S. Son, S. H. Kim, K. Park, K. Choi, and I. C. Kwon,
66. M. M. Gottesman, T. Fojo, and S. E. Bates, Multidrug resistance Self-assembled glycol chitosan nanoparticles for disease-specific
in cancer: Role of ATP-dependent transporters. Nat. Rev. Cancer theranostics. J. Control. Release 193, 202 (2014).
2, 48 (2002). 85. S. J. Yang, F. H. Lin, H. M. Tsai, C. F. Lin, H. C. Chin, J. M. Wong,
67. Y. Patil, T. Sadhukha, L. Ma, and J. Panyam, Nanoparticle-mediated and M. J. Shieh, Alginate-folic acid-modified chitosan nanoparti-
simultaneous and targeted delivery of paclitaxel and tariquidar over- cles for photodynamic detection of intestinal neoplasms. Biomate-
comes tumor drug resistance. J. Control. Release 136, 21 (2009). rials 32, 2174 (2011).
68. H. Meng, M. Liong, T. Xia, Z. Li, Z. Ji, J. I. Zink, and A. E. Nel, 86. H. Z. Zhu, J. H. An, Q. Yao, J. Han, X. T. Li, F. L. Jiang, G. P.
Engineered design of mesoporous silica nanoparticles to deliver Chen, L. N. Peng, Y. S. Li, J. G. Sun, and Z. T. Chen, Chitosan
doxorubicin and P-Glycoprotein siRNA to overcome drug resis- combined with molecular beacon for Mir-155 detection and imag-
tance in a cancer cell line. ACS Nano 4, 4539 (2010). ing in lung cancer. Molecules 19, 14710 (2014).
69. J. Gu, X. Fang, J. Hao, and X. Sha, Reversal of 87. I. Hajdu, G. Trencsenyi, M. Bodnar, M. Emri, G. Banfalvi,
P-glycoprotein-mediated multidrug resistance by CD44 antibody- J. Sikula, T. Marian, J. Kollar, G. Vamosi, and J. Borbely, Tumor-
targeted nanocomplexes for short hairpin RNA-encoding plasmid specific localization of self-assembled nanoparticle PET/MR
DNA delivery. Biomaterials 45, 99 (2015). modalities. Anticancer Res. 34, 49 (2014).

J. Biomed. Nanotechnol. 12, 1585–1603, 2016 1601


Chitosan Nanoparticles in Cancer Treatment Fu et al.

88. G. Zhao, J. Wang, X. Peng, Y. Li, X. Yuan, and Y. Ma, Facile 104. W. I. Choi, J. Y. Kim, C. Kang, C. C. Byeon, Y. H. Kim, and
Solvothermal synthesis of mesostructured FeO/chitosan nanoparti- G. Tae, Tumor regression in vivo by photothermal therapy based on
cles as delivery vehicles for pH-responsive drug delivery and mag- gold-nanorod-loaded, functional nanocarriers. ACS Nano 5, 1995
netic resonance imaging contrast agents. Chem. Asian J. 9, 546 (2011).
(2014). 105. J. F. Liao, W. T. Li, J. R. Peng, Q. Yang, H. Li, Y. Q. Wei,
89. I. C. Sun, J. H. Na, S. Y. Jeong, D. E. Kim, I. C. Kwon, K. Choi, X. N. Zhang, and Z. Y. Qian, Combined cancer photothermal-
C. H. Ahn, and K. Kim, Biocompatible glycol chitosan-coated chemotherapy based on doxorubicin/gold nanorod-loaded polymer-
gold nanoparticles for tumor-targeting CT imaging. Pharm. Res. somes. Theranostics 5, 345 (2015).
31, 1418 (2014). 106. R. Duan, Z. Zhou, G. Su, L. Liu, M. Guan, B. Du, and Q. Zhang,
90. J. H. Ryu, J. H. Na, H. K. Ko, D. G. You, S. Park, E. Jun, H. J. Chitosan-coated gold nanorods for cancer therapy combining chem-
Yeom, D. H. Seo, J. H. Park, S. Y. Jeong, I. S. Kim, B. S. Kim, ical and photothermal effects. Macromol. Biosci. 14, 1160 (2014).
I. C. Kwon, K. Choi, and K. Kim, Non-invasive optical imaging 107. S. Mansouri, P. Lavigne, K. Corsi, M. Benderdour, E. Beaumont,
of cathepsin B with activatable fluorogenic nanoprobes in various and J. C. Fernandes, Chitosan-DNA nanoparticles as non-viral vec-
metastatic models. Biomaterials 35, 2302 (2014). tors in gene therapy: strategies to improve transfection efficacy.
91. Y. A. Shieh, S. J. Yang, M. F. Wei, and M. J. Shieh, Aptamer-based Euro. J. Pharm. Biopharm. 57, 1 (2004).
tumor-targeted drug delivery for photodynamic therapy. ACS Nano 108. R. M. Ramesan and C. P. Sharma, Modification of chitosan
4, 1433 (2010). nanoparticles for improved gene delivery. Nanomedicine 75, 5
92. L. N. Goswami, W. H. White, J. A. Spernyak, M. Ethirajan, (2012).
Y. Chen, J. R. Missert, J. Morgan, R. Mazurchuk, and R. K. Pandey, 109. P. S. Apaolaza, D. Delgado, A. Pozo-Rodriguez, A. R. Gascon, and
Synthesis of tumor-avid photosensitizer-Gd(III)DTPA conjugates: M. A. Solinis, A novel gene therapy vector based on hyaluronic
impact of the number of gadolinium units in T1/T2 relaxivity, acid and solid lipid nanoparticles for ocular diseases. Int. J. Pharm.
intracellular localization, and photosensitizing efficacy. Bioconjug. 465, 413 (2014).
Chem. 21, 816 (2010). 110. R. H. Prabhu, V. B. Patravale, and M. D. Joshi, Polymeric nanopar-
93. S. J. Lee, H. Koo, D. E. Lee, S. Min, S. Lee, X. Chen, Y. Choi, ticles for targeted treatment in oncology: Current insights. Int. J.
J. F. Leary, S. Y. Jeong, I. C. Kwon, K. Kim, and K. Choi, Nanomed. 10, 1001 (2015).
Tumor-homing photosensitizer-conjugated glycol chitosan nanopar- 111. J. O. You, Y. C. Liu, and C. A. Peng, Efficient gene transfection
ticles for synchronous photodynamic imaging and therapy based on using chitosan-alginate core–shell nanoparticles. Int. J. Nanomed.
cellular on/off system. Biomaterials 32, 4021 (2011). 1, 173 (2006).
94. Y. Sun, Z. Chen, X. Yang, P. Huang, X. Zhou, and X. Du, Mag- 112. J. Halder, A. A. Kamat, C. N. Landen, Jr., L. Y. Han, S. K.
netic chitosan nanoparticles as a drug delivery system for targeting Lutgendorf, Y. G. Lin, W. M. Merritt, N. B. Jennings, A. Chavez-
photodynamic therapy. Nanotechnology 20, 135102 (2009). Reyes, R. L. Coleman, D. M. Gershenson, R. Schmandt, S. W.
95. Q. T. Chen, X. Wang, F. H. Chen, Q. B. Zhang, B. Dong, H. Yang, Cole, G. Lopez-Berestein, and A. K. Sood, Focal adhesion kinase
Deliveredofby
G. X. Liu, and Y. M. Zhu, Functionalization Ingentalumi-
upconverted to: Main CIDtargeting
is 80004805
using in (JPP)
vivo short interfering RNA delivery in neu-
IP: 146.185.205.29
nescent NaYF4: Yb/Er nanocrystals by folic acid-chitosan con- On: Wed, 07 tral liposomes 04:41:09
Dec 2016 for ovarian carcinoma therapy. Clin. Cancer Res.
Copyright:
jugates for targeted lung cancer cell imaging. American
J. Mater. Chem. Scientific Publishers
12, 4916 (2006).
21, 7661 (2011). 113. H. D. Han, L. S. Mangala, J. W. Lee, M. M. K. Shahzad, H. S.
96. G. Chen, H. Qiu, P. N. Prasad, and X. Chen, Upconversion nanopar- Kim, D. Shen, E. J. Nam, E. M. Mora, R. L. Stone, C. Lu, S. J.
ticles: Design, nanochemistry, and applications in theranostics. Lee, J. W. Roh, A. M. Nick, G. Lopez-Berestein, and A. K. Sood,
Chem. Rev. 114, 5161 (2014). Targeted gene silencing using RGD-labeled chitosan nanoparticles.
97. V. J. Pansare, S. Hejazi, W. J. Faenza, and R. K. Prud’homme, Clin. Cancer Res. 16, 3910 (2010).
Review of long-wavelength optical and NIR imaging materials: 114. F. Q. Hu, W. W. Chen, M. D. Zhao, H. Yuan, and Y. Z. Du,
Contrast agents, fluorophores, and multifunctional nano carriers. Effective antitumor gene therapy delivered by polyethylenimine-
Chem Mater. 24, 812 (2012). conjugated stearic acid-chitosan oligosaccharide micelles. Gene
98. H. Guo and S. Sun, Lanthanide-doped upconverting phosphors for Ther. 20, 597 (2013).
bioassay and therapy. Nanoscale 4, 6692 (2012). 115. C. He, L. Yin, Y. Song, C. Tang, and C. Yin, Optimization of mul-
99. L. Cheng, K. Yang, S. Zhang, M. Shao, S. T. Lee, and Z. Liu, tifunctional chitosan-siRNA nanoparticles for oral delivery appli-
Highly-sensitive multiplexed in vivo imaging using PEGylated cations, targeting TNF- silencing in rats. Acta Biomater. 17, 98
upconversion nanoparticles. Nano Res. 3, 722 (2010). (2015).
100. Z. Gu, L. Yan, G. Tian, S. Li, Z. Chai, and Y. Zhao, Recent 116. A. V. Nascimento, A. Singh, H. Bousbaa, D. Ferreira, B. Sarmento,
advances in design and fabrication of upconversion nanoparti- and M. M. Amiji, Mad2 checkpoint gene silencing using epidermal
cles and their safe theranostic applications. Adv. Mater. 25, 3758 growth factor receptor-targeted chitosannanoparticles in non-small
(2013). cell lung cancer model. Mol Pharma. 11, 3515 (2014).
101. U. Gneveckow, A. Jordan, R. Scholz, V. Brub, N. Waldofner, 117. L. Han, J. Zhao, X. Zhang, W. Cao, X. Hu, G. Zou, X. Duan,
J. Ricke, A. Feussner, B. Hildebrandt, B. Rau, and P. Wust, and X. J. Liang, Enhanced siRNA delivery and silencing gold-
Description and characterization of the novel hyperthermia- and chitosan nanosystem with surface charge-reversal polymer assem-
thermoablation-system MFH® 300F for clinical magnetic fluid bly and good biocompatibility. ACS Nano 28, 7340 (2012).
hyperthermia. Med. Phys. 31, 1444 (2004). 118. E. Salva, S. O. Turan, F. Eren, and J. Akbuga, The enhancement
102. F. R. Li, W. H. Yan, Y. H. Guo, H. Qi, and H. X. Zhou, Preparation of gene silencing efficiency with chitosan-coated liposome formu-
of carboplatin-Fe@C-loaded chitosan nanoparticles and study on lations of siRNAs targeting HIF-1 and VEGF. Int. J. Pharm.
hyperthermia combined with pharmacotherapy for liver cancer. Int. 478, 147 (2015).
J. Hypertherm. 25, 383 (2009). 119. M. M. Farid, R. M. Hathout, M. Fawzy, and K. Abou-Aisha, Silenc-
103. J. You, R. Zhang, G. Zhang, M. Zhong, Y. Liu, C. S. Van ing of the scavenger receptor (Class B-Type 1) gene using siRNA-
Pelt, D. Liang, W. Wei, A. K. Sood, and C. Li, Photothermal- loaded chitosan nanaoparticles in a HepG2 cell model. Colloi. Surf.
chemotherapy with doxorubicin-loaded hollow gold nanospheres: B 123, 930 (2014).
A platform for near-infrared light-trigged drug release. J. Control. 120. L. Han, J. Zhao, J. Liu, X. L. Duan, L. H. Li, X. F. Wei, Y. Wei,
Release 158, 319 (2012). and X. J. Liang, A universal gene carrier platform for treatment

1602 J. Biomed. Nanotechnol. 12, 1585–1603, 2016


Fu et al. Chitosan Nanoparticles in Cancer Treatment

of human prostatic carcinoma by p53 transfection. Biomaterials 135. C. Chen, J. L. Zhou, X. Han, F. Song, X. L. Wang, and Y. Z. Wang,
35, 3110 (2014). A prodrug strategy based on chitosan for efficient intracellular anti-
121. Z. Chen, L. Zhang, Y. He, Y. Shen, and Y. Li, Enhanced shRNA cancer drug delivery. Nanotechnology 25, 255101 (2014).
delivery and ABC G2 silencing by charge-reversible layered 136. B. Lu, D. Huang, H. Zheng, Z. Huang, P. Xu, H. Xu, Y. Yin, X. Liu,
nanocarriers. Small 11, 952 (2015). D. Li, and X. Zhang, Preparation, characterization, and in vitro
122. J. H. Kim, Y. S. Kim, S. Kim, J. H. Park, K. Kim, K. Choi, efficacy of O-carboxymethyl chitosan conjugate of melphalan. Car-
H. Chung, S. Y. Jeong, R. W. Park, I. S. Kim, and I. C. Kwon, bohydr Polym. 98, 36 (2013).
Hydrophobically modified glycol chitosan nanoparticles as carriers 137. P. Sanpui, A. Chattopadhyay, and S. S. Ghosh, Induction of apop-
for paclitaxel. J. Control. Release 111, 228 (2006). tosis in cancer cells at low silver nanoparticle concentrations using
123. F. Li, J. Li, X. Wen, S. Zhou, X. Tong, P. Su, H. Li, and D. Shi, chitosan nanocarrier. ACS Appl. Mater. Interf. 3, 218 (2011).
Anti-tumor activity of paclitaxel-loaded chitosan nanoparticles: An 138. M. Amidi, E. Mastrobattista, W. Jiskoot, and W. E. Hennink,
in vitro study. Mater. Sci. Engine. C 29, 2392 (2009). Chitosan-based delivery systems for protein therapeutics and anti-
124. W. Wei, P. P. Lv, X. M. Chen, Z. G. Yue, Q. Fu, S. Y. Liu, H. Yue, gens. Adv. Drug Deliv. Rev. 62, 59 (2010).
and G. H. Ma, Codelivery of mTERT siRNA and paclitaxel by 139. S. Lien and H. B. Lowman, Therapeutic peptides. Trends Biotech-
chitosan-based nanoparticles promoted synergistic tumor suppres- nol. 21, 556 (2003).
sion. Biomaterials 34, 3912 (2013). 140. Z. Antosova, M. Mackova, V. Kral, and T. Macek, Therapeutic
125. Z. Chen, L. Zhang, Y. Song, J. He, L. Wu, C. Zhao, Y. Xiao, W. Li, application of peptides and proteins: Parenteral forever? Trends
B. Cai, H. Cheng, and W. Li, Hierarchical targeted hepatocyte mito- Biotechnol. 27, 628 (2009).
chondrial multifunctional chitosan nanoparticles for anticancer drug 141. S. Frokjaer and D. E. Otzen, Protein drug stability: A formulation
delivery. Biomaterials 52, 240 (2015). challenge. Nat. Rev. Drug Discov. 4, 298 (2005).
126. V. Nipun Babu and S. Kannan, Enhanced delivery of baicalein 142. P. Fonte, J. C. Andrade, V. Seabra, and B. Sarmento, Chitosan-
using cinnamaldehyde cross-linked chitosan nanoparticle inducing based nanoparticles as delivery systems of therapeutic proteins.
apoptosis. Int. J. Biol. Macromol. 51, 1103 (2012). Therapeutic Proteins: Methods and Protocols. Methods in Molecu-
127. X. P. Liu, S. T. Zhou, X. Y. Li, X. C. Chen, X. Zhao, Z. Y. Qian, lar Biology 899, 471 (2012).
L. N. Zhou, Z.Y. Li, Y. M. Wang, Q. Zhong, T. Yi, Z. Y. Li, 143. M. George and T. E. Abraham, Polyionic hydrocolloids for the
X. He, and Y. Q. Wei, Anti-tumor activity of N -trimethyl chitosan- intestinal delivery of protein drugs: Alginate and chitosan-a review.
encapsulated camptothecin in a mouse melanoma model. J. Exper. J. Control. Release 114, 1 (2006).
Clin. Cancer Res. 29, 76 (2010). 144. C. J. Roberts, Therapeutic protein aggregation: Mechanisms,
128. L. H. Chuah, C. J. Roberts, N. Billa, S. Abdullah, and R. Rosli, design, and control. Trends Biotechnol. 32, 372 (2014).
Cellular uptake and anticancer effects of mucoadhesive curcumin- 145. M. Werle and A. Bernkop-Schnurch, Strategies to improve plasma
containing chitosan nanoparticles. Colloi. Surf. B 116, 228 half life time of peptide and protein drugs. Amino Acids 30, 351
(2014). (2006).
129. L. Bu, L. C. Gan, X. Q. Guo, F. Delivered
Z. Chen, Q. bySong,
Ingenta to: Main146.
Q. Zhao, CIDY. is
Shi80004805
and G. Huang, (JPP)
Recent developments of biodegradable and
X. J. Gou, S. X. Hou, and Q. Yao, IP:Trans-resveratrol
146.185.205.29 loadedOn:
chi- Wed, 07biocompatible
Dec 2016 materials
04:41:09 based micro/nanoparticles for delivering
Copyright:
tosan nanoparticles modified with biotin and American
avidin to target hepatic Scientific Publishers
macromolecular therapeutics. Crit. Rev. Ther. Drug Carrier Syst.
carcinoma. Int. J. Pharm. 452, 355 (2013). 26, 29 (2009).
130. M. Guan, Y. Zhou, Q. L. Zhu, Y. Liu, Y. Y. Bei, X. N. Zhang, and 147. F. Andrade, F. Antunes, A. V. Nascimento, S. B. da Silva, J. das
Q. Zhang, N -trimethyl chitosan nanoparticle-encapsulated lactosyl- Neves, D. Ferreira, and B. Sarmento, Chitosan formulations as car-
norcantharidin for liver cancer therapy with high targeting efficacy. riers for therapeutic proteins. Curr. Drug Discov. Technol. 8, 157
Nanomedicine 8, 1172 (2012). (2011).
131. C. S. Hu, C. H. Chiang, P. D. Hong, and M. K. Yeh, Influence of 148. M. Thanou, B. I. Florea, M. W. Langemeyer, J. C. Verhoef,
charge on FITC-BSA-loaded chondroitin sulfate-chitosan nanopar- and H. E. Junginger, N -trimethylated chitosan chloride (TMC)
ticles upon cell uptake in human Caco-2 cell monolayers. Int. J. improves the intestinal permeation of the peptide drug buserelin in
Nanomed. 7, 4861 (2012). vitro (Caco-2 cells) and in vivo (rats). Pharm. Res. 17, 27 (2000).
132. Y. Xu, L. Wang, Y. K. Li, and C. Q. Wang, Oxidation and 149. J. H. Kim, Y. S. Kim, K. Park, E. Kang, S. Lee, H. Y. Nam,
pH responsive nanoparticles based on ferrocene-modified chi- K. Kim, J. H. Park, D. Y. Chi, R. W. Park, I. S. Kim, K. Choi, and
tosan oligosaccharide for 5-fluorouracil delivery. Carbohydr Polym. I. C. Kwon, Self-assembled glycol chitosan nanoparticles for the
114, 27 (2014). sustained and prolonged delivery of antiangiogenic small peptide
133. J. H. Kim, Y. S. Kim, K. Park, S. Lee, H. Y. Nam, K. H. Min, drugs in cancer therapy. Biomaterials 29, 1920 (2008).
H. G. Jo, J. H. Park, K. Choi, S. Y. Jeong, R. W. Park, I. S. Kim, 150. J. H. Park, S. Kwon, J. O. Nam, R. W. Park, H. Chung, S. B. Seo,
K. Kim, and I. C. Kwon, Antitumor efficacy of cisplatin-loaded I. S. Kim, I. C. Kwon, and S. Y. Jeong, Self-assembled nanopar-
glycol chitosan nanoparticles in tumor-bearing mice. J. Control ticles based on glycol chitosan bearing 5-cholanic acid for RGD
Release. 127, 41 (2008). peptide delivery. J. Control. Release 95, 579 (2004).
134. A. Babu, Q. Wang, R. Muralidharan, M. Shanker, A. Munshi, 151. J. H. Kim, S. M. Bae, M. H. Na, H. Shin, Y. J. Yang, K. H.
and R. Ramesh, Chitosan coated polylactic acid nanoparticle- Min, K. Y. Choi, K. Kim, R. W. Park, I. C. Kwon, B. H. Lee,
mediated combinatorial delivery of cisplatin and siRNA/plasmid A. S. Hoffman, and I. S. Kim, Facilitated intracellular delivery of
DNA chemosensitizes cisplatin-resistant human ovarian cancer peptide-guided nanoparticles in tumor tissues. J. Control. Release
cells. Mol. Pharm. 11, 2720 (2014). 157, 493 (2012).

J. Biomed. Nanotechnol. 12, 1585–1603, 2016 1603


KANSER TEDAVİSİNDE FONKSİYONEL KİTOSAN NANOPARTİKÜLLERİ
Shaozhi Fu, Jiyi Xia ve Jingbo Wu tarafından yazılan bu makalede kitosanın kanser
tedavilerinde nasıl ve hangi amaçla kullanıldığına dair bilgiler verilmiştir.
Kanserin insan sağlığını nasıl tehdit ettiğini bilmeyen yoktur.Öyle ki 2030 yılına kadar dünya
çapında 12 milyon insanın kanser hastalığı yüzünden öleceği ön görülüyor. Özellikle Çin gibi
gelişmiş ve büyük ülkelerde çevre kirliliği yüzünden yıldan yıla kanser hastalığı ve ölümü
artmakta.Kanser tedavilerinde en çok kemoterapi gibi tedavi yöntemleri kullansa da bu
tedavi yöntemlerinin çok ciddi yan etkileri bulunur.Yeni kanser tedavisi keşfetme sürecinde
malzeme bilimi sayesinde önemli gelişmeler yaşanmıştır.Kitosan da bu tedavi
yöntemlerinden biridir.Doğal bir polisakkarit olan kitosan büyük ölçekte kitinin alkali N-
deasetilasyonu ile üretilir.Kitin bol bir polimerdir ve kitin kabuklu hayvanların kabuğunda
bulunur.Kitosan çeşitli kemoterapötik ilaçların dağıtımı için ilaç taşıyıcısı olarak yaygın
kullanılsa da sadece ilaç yükleme aracı görevi görür ve kanser tedavisinde rol oynamaz.Bu
nedenle başka bir şey önerildi.Kitosan omurgasında ki zengin reaktif gruplar sayesinde bazı
hidrofobik antikanser ilacı hidrofilik kitosan zincirlerine doğrudan konjuge edilebilir ve bu da
iyi bir sonuç verir.

You might also like