Interchitosan4 PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Carbohydrate Polymers 222 (2019) 115004

Contents lists available at ScienceDirect

Carbohydrate Polymers
journal homepage: www.elsevier.com/locate/carbpol

Review

Advances in chitosan-based nanoparticles for oncotherapy T


a,b a,b,⁎ a,b a,b a,b a,b,⁎
Enhui Zhang , Ronge Xing , Song Liu , Yukun Qin , Kecheng Li , Pengcheng Li
a
Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
b
Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China

A R T I C LE I N FO A B S T R A C T

Keywords: Chitosan has attracted considerable attention as an anti-tumor drug carrier material in recent years, which is due
Chitosan to its biocompatibility and biodegradability, as well as the simple and mild preparing techniques of drug-loaded
Nanoparticle nanoparticles. Chitosan-based nanoparticles can deliver various anti-tumor agents to specific tumor tissues by
Anti-tumor passive and active targeting mechanisms, including traditional chemotherapeutic agents, DNA or siRNA, pro-
Drug delivery
teins, photosensitizers and so on. In this review, we summarized the factors affecting the anti-tumor efficacy of
chitosan-based nanoparticles, to aid exploring the function-structure relationship. The recent studies on chit-
osan-based nanoparticles for oncotherapy were highlighted, including their structures, properties and phar-
macological effects. Finally, we offered our perspectives on the challenges and future development of this area.

1. Introduction rabbits had no obvious effects in 65 days (Kean & Thanou, 2010). In a
phase II clinical trial, percutaneous injection of holmium-166/chitosan
Chitosan, a unique alkaline polysaccharide in nature, is derived complex for the treatment of hepatocellular carcinoma on patients in-
from chitin by a process of deacetylation, comprising of β-(1→4)-linked duced very little discomfort and disruption of daily activities for pa-
D-glucosamine and N-acetyl-D-glucosamine units. It was firstly prepared tients (Kim et al., 2006). From most studies, chitosan exhibited non- or
by Charles Rouget in 1859, who boiled chitin in the presence of con- minimal toxicity, which made it widely regarded as a safe material in
centrated potassium hydroxide to obtain chitosan (Rouget, 1859). The drug delivery (Smith, Perelman, & Hinchcliffe, 2014).
modified chitin was named “chitosan” after 35 years later by Felix One of the most essential properties of chitosan is the cationic
Hoppe Seyler (Hoppe-Seyler, 1894) and the structure of chitosan was nature, which exerts advantages as a desirable drug carrier due to the
finally resolved in 1950 (Baldrick, 2010). In the wake of development enhanced adhesion to the negatively charged mucosal surface by elec-
of drug delivery systems in 1970s, chitosan continuously attracted the trostatic interaction, resulting in the improvement on drug inter-
attention as a promising candidate for the drug carrier material due to nalization into targeted cells (He, Davis, & Illum, 1998; Lee et al., 2014;
its favorable properties such as biocompatibility, biodegradability, non- Narayanan, Jayakumar, & Chennazhi, 2014). On the other hand, the
toxicity and low immunogenicity (Garcia-Fuentes & Alonso, 2012). cationic property of chitosan also makes it potential to complex abun-
The safety is always the priority concern for a qualified pharma- dant anionic nucleic acid, protecting from degradation by nuclease in
ceutical excipient, and chitosan is commonly considered to be a safe serum, achieving efficient gene therapy (Rudzinski & Aminabhavi,
and biocompatible material. The oral administration of chitosan with 2010).
dose of 6.75 g for 8 weeks induced no observable effects on serum vi- However, chitosan is only soluble in an aqueous acidic medium but
tamin, carotenes levels or other safety parameters in human volunteers not at neutral pH, which is a major hurdle for its application. The
(Tapola, Lyyra, Kolehmainen, Sarkkinen, & Schauss, 2008). Further- abundant amino and hydroxyl groups on chitosan backbone represent
more, intravenous injection of chitosan with 7.1–8.6 mg/kg/day in target moieties for chemical modifications to improve the aqueous

Abbreviations: Mw, molecular weight; DD, degree of deacetylation; PEG, poly(ethylene glycol); FITC, fluorescein isothiocyanate; EPR, enhanced permeability and
retention; APBA, N-3-acrylamidophenylboronic acid; MRP, multidrug resistance-associated protein; TPP, tripolyphosphate; mPEG, methoxy PEG; EGCG, epigallo-
catechin-3-gallate; Ce6, chlorin e6; CT, X-ray computed tomography; MR, magnetic resonance; PAMAM, polyamidoamine; ZWC, zwitterionic chitosan; VEGF,
vascular endothelial growth factor; MDR, multi drug resistance; P-gp, P-glycoprotein; mTERT, mouse telomerase reverse transcriptase; PEI, poly(ethylene imine);
PLR, poly-L-arginine; PLL, poly-L-lysine; PAH-Cit, poly(allylamine hydrochloride)-citraconic anhydride; γ-PGA, poly(γ-glutamic acid); PpIX, protophorphyrin IX;
DEAP, 3-diethylaminopropyl isothiocyanate; CpG, cytosine-guanine; IL-12, interleukin-12

Corresponding authors at: Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences,
Qingdao 266071, China.
E-mail addresses: xingronge@qdio.ac.cn (R. Xing), pcli@qdio.ac.cn (P. Li).

https://doi.org/10.1016/j.carbpol.2019.115004
Received 21 February 2019; Received in revised form 27 May 2019; Accepted 17 June 2019
Available online 19 June 2019
0144-8617/ © 2019 Elsevier Ltd. All rights reserved.
E. Zhang, et al. Carbohydrate Polymers 222 (2019) 115004

Fig. 1. Structural features of chitosan and their influences on chitosan-based nanoparticles for oncotherapy.

solubility, as well as endow chitosan with new functionalities, such as higher Mw (> 200 kDa) was better (Huang, Fong, Khor, & Lim, 2005;
targeted drug delivery and environment-sensitive drug release, enhan- Weecharangsan et al., 2008) but lower Mw (< 10 kDa) had success in
cing the therapeutic effects and reducing side effects. The most common other cases (Köping-Höggård et al., 2004; Sato, Ishii, & Okahata, 2001).
chitosan derivatives used for drug carriers include thiolated chitosan, There is a subtle balance between stability and dissociation inside cells
trimethyl chitosan, carboxymethyl chitosan, glycol chitosan and so on influenced by chitosan Mw. Higher Mw increases the stability of chit-
(Sarmento & Bruno, 2012). osan/DNA complexes but delayed their dissociation and subsequent
In this review, we first described the influence of chitosan structural gene expression in cells, while lower Mw has the opposite effects. The
features on anti-tumor efficacy of nanoparticles and the characteriza- balance also exists in chitosan/siRNA complexes, but there are few
tion of medical grade chitosan. Furthermore, we highlighted recent reports on successful delivery of siRNA based on chitosans with low Mw
research advances in anti-tumor chitosan nanoparticles, categorized by (< 10 kDa). A study reported that chitosan/siRNA nanoparticles with
the types of delivered drugs. Finally, we made a brief comment on the low Mw (below 10 kDa) had almost no gene silencing efficacy, while
future development and challenges of chitosan nanoparticles for on- those with higher Mw (114 and 170 KDa) exhibited effective silencing
cotherapy (Tables 1–3). efficacy, which was comparable to commercial reagents (Liu et al.,
2007). Furthermore, complete binding of siRNA into chitosan nano-
2. Factors affecting the efficacy of chitosan-based nanoparticles particles (weight ratio of chitosan and siRNA, 50:1) could be observed
with chitosans Mw of 25 kDa at least but not those with Mw below
The structural features of chitosan and its derivatives, such as mo- 10 kDa (Fernandes et al., 2012).
lecular weight (Mw), degree of deacetylation (DD), degree and site of The DD of chitosan determines the positive charge density and the
substitutions, have great impact on physiochemical properties as well binding capacity with DNA/siRNA. Higher DD is conducive to escape
as the anti-tumor efficacy of chitosan-based nanoparticles (Fig. 1). from the endo-lysosomal compartment for transfection (Huang et al.,
Choosing appropriate chitosan is a priority issue for rational designed 2005; Liu et al., 2007). DD of more than 46% is a basic requirement for
nanoparticles. The Mw of commercial chitosan ranges from 10 kDa to 2 obtaining stable chitosan-based DNA complexes (Mao, Sun, & Kissel,
000 kDa and the DD is between 60% and 100%. In 2003, Huang et al. 2010). Kiang et al. reported the disparity of in vitro and in vivo trans-
firstly conducted a comprehensive assessment of effects of Mw and DD fection efficiency of chitosan/DNA complexes with large Mw chitosan
on cellular uptake and cytotoxicity of chitosan nanoparticles. The Mw (390 kDa) and DD of 90%, 70% and 62%. The decrease of DD induced
of chitosan in this study ranged from 10 kDa to 213 kDa and DD of 46% the decrease of in vitro gene expression due to the instability of com-
to 88%. The cellular uptake results showed positive correlation with plexes, but this instability, in turn, increased the in vivo gene expression
zeta potential of nanoparticles, and the uptake ratios reduced by 26% (Kiang, Wen, Lim, & Leong, 2004). As for siRNA, Malmo et al. proved
and 41% with the decrease of Mw and DD, respectively. The decrease of that fully N-acetylated chitosan had superior silencing efficacy com-
DD also attenuated the cytotoxicity of nanoparticles against A549 cells, pared to partially N-acetylated chitosan (Malmo, Sørgård, Vårum, &
but the impact of Mw was smaller (Huang, Khor, & Lim, 2004). Fur- Strand, 2012). Most cases suggested that DD of more than 80% is
thermore, Park et al. evaluated the effect of Mw (20 kDa, 100 kDa, mandatory for efficient delivery of DNA/siRNA and transfection effi-
250 kDa) on the physiochemical properties and in vivo activities of ciency (Alameh et al., 2018; Buschmann et al., 2013; Liu et al., 2007;
glycol chitosan nanoparticles. The size and zeta potential did not differ Ragelle, Vandermeulen, & Préat, 2013).
significantly, but nanoparticles with Mw 250 kDa exhibited longer cir- In addition, the degree and site of substitutions grafted onto chit-
culation time and better tumor selectivity (Park et al., 2007). On the osan are also important factors in synthesis of chitosan nanoparticles. Li
contrary, nanoparticles fabricated by amphiphilic chitosan derivatives and co-workers synthesized a series of N-deoxycholic acid-N,O-glycol
with low Mw (5 kDa, 10 kDa, 20 kDa) also showed desirable in vitro chitosans with different degree of substitution of glycol and deoxycholic
cytotoxicity against HepG2 cells. However, no sufficient in vivo tests acid group. The increase of deoxycholic acid, the hydrophobic group,
were reported to confirm the efficacy of nanoparticles based on chit- resulted in the increase of particle size, entrapment efficiency and drug
osan with low Mw (Zhang, Huo, Zhou, Yu, & Wu, 2009). Based on loading content of paclitaxel significantly (Li et al., 2010). Furthermore,
above studies, Mw is not a key factor that influences the physiochemical it was found that the content of hydrophobic groups could influence the
characteristics of chitosan nanoparticles, but relatively high Mw en- stability and deformability of nanoparticles based on N-5β-cholanic
sures the longer circulation time of nanoparticles in bloodstream, which acid-glycol chitosan (CNPs). Despite that the prepared nanoparticles
subsequently guarantees the high tumor selectivity. High DD produces were over 300 nm in size, they could deform and pass through the
high surface charge density of nanoparticles, resulting in enhanced 0.2 μm pore filter, as shown in Fig. 2. CNPs with smaller wt.% of 5β-
cellular uptake and anti-tumor efficacy. cholanic acid exhibited excellent deformability but unsatisfactory sta-
As for DNA, the binding ability of chitosan with DNA and trans- bility in serum. Thus, the balanced stability and deformability of na-
fection efficiencies changes along with chitosan Mw. However, di- noparticles ensured the superior in vivo tumor targeting efficiency.
vergent results were observed in exploring the influence of Mw on Amongst, nanoparticles with 23 wt.% of 5β-cholanic acid exhibited the
transfection efficiency of chitosan/DNA complexes. In some studies, highest tumor accumulation (Na et al., 2012). Moreover, the site of

2
E. Zhang, et al. Carbohydrate Polymers 222 (2019) 115004

Fig. 2. The deformability test of Cy5.5-labeled


CNPs with different wt.% of 5β-cholanic acid to
glycol chitosan by measuring the fluorescence
intensity before and after filtration at different
pore sizes (A). The fluorescence intensities be-
fore and after filtration (B). The rigid and non-
deformable polystyrene nanoparticles (PSNPs)
were employed as control. Reproduced with
permission from (Na et al., 2012). Copyright
2012, Elsevier. Abbreviation: GC, glycol chit-
osan.

carboxymethylation on chitosan is also closely related to the anti-tumor 4. Applications in drug delivery
activity and cellular uptake of carboxymethyl chitosan-based nano-
particles. O-carboxymethylation showed superior activity to N-carbox- 4.1. Chitosan-based systems as carriers of chemotherapeutic agents
ymethylation and N,O-carboxymethylation, which might be attributed
to the reservation of amino groups that facilitate cellular uptake Currently, chemotherapy is one of the most essential tools for on-
(Zhang, Xing et al., 2017). Besides, as for nanoparticles prepared by cotherapy in clinic, which is usually used to shrink the tumor size be-
chitosan and poly(ethylene glycol) (PEG) derivatives, the addition of fore or after surgery. Common chemotherapeutic agents, including
excessive PEG could decrease the zeta potential due to the shielding of doxorubicin, paclitaxel, docetaxel, cisplatin, camptothecin and so on,
nanoparticle surface by PEG (Huo et al., 2012). have exhibited stronger cytotoxicity against tumor cells than normal
The characteristics of chitosan and its derivatives, such as Mw, DD, cells and made enormous contributions in oncotherapy for the past
degree and site of substitutions, have remarkable impact on the efficacy decades. However, these drugs still suffer severe drawbacks, such as
of chitosan-based nanoparticles. It is important to note that these fac- dose-dependence and occasional lethal cardiomyopathy of doxorubicin,
tors are not independent but exert combined influence on the nano- various severe side effects caused by usage of organic solvent in taxane
particles. Therefore, choosing proper chitosan as nanocarriers needs injections. Numerous studies have been conducted to solve these pro-
comprehensive consideration on the relationship of above factors with blems by incorporating chemotherapeutic agents with various chitosan-
the properties of payloads, preparing method of nanoparticles, target based nanoparticles. According to the difference in preparing method,
disease and so on. the nanoparticles can be divided into three categories: self-assembled
nanoparticle, ionic cross-linked nanoparticle and polyelectrolyte com-
plex. Their structures are depicted in Fig. 3.
3. Characterization of medical grade chitosans

Chitosan is a common medical material and pharmaceutical ex- 4.1.1. Self-assembled nanoparticles
cipient extracted from natural sources, so a standard for medical grade Self-assembled nanoparticles have a micelle-like structure with a
chitosan is necessary. Establishing the standard will ensure the batch- hydrophobic inner core and a hydrophilic out shell, which are fabri-
to-batch consistency of production and therapeutic efficacy as well as cated by amphiphilic chitosan derivatives (Fig. 3). In aqueous phase,
safety of chitosan materials. Different pharmacopeias and societies the hydrophobic chains spontaneously aggregated together to form a
provided standard guides for chitosan or its salts. United States reservoir for both soluble and sparingly soluble drugs, and hydrophilic
Pharmacopoeia (USP) regards the DD of chitosan between 70.0% and chains served as a shell surrounded the core being exposed to the
95.0% as acceptance criteria and the residue on ignition should be not aqueous phase.
more than 1.0%. There are also requirements on the limit of lead, Hydrophobically modified glycol chitosan is one of the most com-
mercury, chromium, nickel, cadmium, arsenic as well as iron. monly used derivatives to fabricate self-assembled nanoparticles. Kwon
Furthermore, the level of bacterial endotoxins should meet the re- and co-workers synthesized a series of hydrophobic moieties con-
quirements of related dosage form monographs using chitosan (United jugated glycol chitosans as nanocarriers, including 5β-cholanic acid,
States Pharmacopoeia Convention, 2016). Recent studies showed that fluorescein isothiocyanate (FITC), deoxycholic acid, palmitoyl group
the endotoxin content played an important role in immune responses, and so on. The encapsulated drugs included both soluble and insoluble
which was related to Mw, DD of chitosan, and endotoxin structure molecules, such as cisplatin, camptothecin, paclitaxel, docetaxel and so
(Gjoseva et al., 2018; Ravindranathan, Koppolu, Smith, & Zaharoff, on (Bei et al., 2014; Hwang, Kim, Kwon, & Kim, 2008; Kim et al., 2005,
2016). Besides, American Society for Testing and Materials (ASTM) also 2008; Min et al., 2008). A recent study demonstrated that glycol chit-
provide a standard guide for chitosan salt as starting materials used for osan-5β-cholanic acid-based nanoparticles have longer circulation time
biomedical and tissue-engineered medical product applications. The and stronger liver tumor targeting ability than liposomes, polystyrene
guide lists a series of detection methods for DD, Mw, viscosity, dry nanoparticles with similar diameters (Na et al., 2016). Furthermore,
matter content, ash content, insolubles. Similar to USP, the content of conjugating 2-(4-(vinylbenzyloxy)-N,N-diethylnicotinamide) oligomers
endotoxin is also listed as one of the major impurities of chitosan with glycol chitosan could significantly increase the drug loading
products. Other impurities include protein content, heavy metal content content up to 24.2 wt.% of paclitaxel, and the in vivo anti-tumor activity
and microorganisms (American Society for Testing & Materials, 2011). was significantly stronger than the free drug and Abraxane® (Koo et al.,
2013). The conjugation of octylamine could increase the drug loading
content over 30%, and the tumor inhibition rate was 90.3% compared

3
E. Zhang, et al. Carbohydrate Polymers 222 (2019) 115004

with paclitaxel of 58.7% in MDA-MB-231 tumor bearing mice (Huo

(Wang, Zhen et al., 2013)

(Lee, Termsarasab et al.,


(Hsiao, Mu et al., 2015)
et al., 2016). For further improvement of selective delivery of drugs, a

(Huang et al., 2015)


variety of ligands were often introduced in glycol chitosan nano-

(Chen et al., 2018)

(Yang et al., 2017)


(Huo et al., 2012)

(Huo et al., 2016)

(Liu & Yeo, 2013)


(Tsai et al., 2018)
(Min et al., 2008)

(Zhu et al., 2018)

(He & Yin, 2017)


(Hu et al., 2015)

(Jia et al., 2014)


particles, such as somatostatin (Huo et al., 2012), peptides (Key et al.,
2016; Zhang et al., 2019), folic acid (Yu et al., 2013) and so on. Besides,
Reference

carboxymethyl chitosan is another soluble chitosan derivative and

2017)
widely studied as drug carriers. As with glycol chitosan, carboxymethyl
chitosan can self-assemble into nanoparticles after hydrophobically
modified, such as quercetin (Wang et al., 2014), linoleic acid (Tan &
in vitro and in vivo
in vitro and in vivo
in vitro and in vivo
in vitro and in vivo

in vitro and in vivo

in vitro and in vivo


in vitro and in vivo

in vitro and in vivo


in vitro and in vivo
in vitro and in vivo

in vitro and in vivo


in vitro and in vivo
Biological status

Liu, 2009), 4-mercaptobenzoic acid (Gao, Liu et al., 2014) or hydro-


phobic drug, methotrexate (Wang et al., 2011). Unlike glycol chitosan,
the surfaces of carboxymethyl chitosan nanoparticles were negative
in vitro

in vitro

in vitro
charged, which also showed high cellular uptake and strong anti-tumor
efficacy. Huang et al. synthesized hexanoyl-grafted carboxymethyl
chitosan as drug carriers coated with an anti-EGFR antibody layer for
both oral and intravenous delivery of paclitaxel

chemo-photodynamic-gene therapy, bioimaging


high redox sensitivity of drug release in tumor

delivery of demethoxycurcumin. The prepared nanoparticles exhibited


overcome microenvironment mediated drug
high hydrophobic drug loading, targeting

8-fold reduction of tumor volume in A549 xenograft lung tumor mice

surface charge reversible nanoparticles


targeted chemo-photodynamic therapy

compared with the control group (Huang et al., 2015).


enhance the penetration into tumor

co-delivery of doxorubicin and its

Amphiphilic chitosan-grafted copolymers have also been in-


enhance passive targeting ability
enhance active targeting ability
enhance active targeting ability

overcome multidrug resistance

synergistical anti-tumor effect

vestigated to fabricate self-assembled nanoparticles. Hsiao et al. syn-


delivery to glioblastoma cells
redox-sensitive drug release
high drug loading content,

thesized an amphiphilic hexanoyl-chitosan-PEG copolymer, which was


used to coat paclitaxel-loaded and chlorotoxin-grafted iron oxide na-
Anti-tumor strategy

noparticles for targeted delivery of paclitaxel to glioblastoma cells.


chemosensitizer

Chlorotoxin enhanced cellular uptake of nanoparticles in U-118 MG


cells and therefore improved the anti-tumor activity (Hsiao, Mu,
resistance

Stephen, Fang, & Zhang, 2015). Furthermore, Jiang et al. designed a


delicate nanoparticle by polymerization of N-3-acrylamidophe-
nylboronic acid (APBA) in the presence of chitosan, then forming am-
cancer associated fibroblasts,

NIH/3T3 cells, SKOV-3 cells

phiphilic copolymers by the coordination between boronic acid of poly-


SH-SY5Y cells, H-22 cells

MCF-7 cells, EMT-6 cells

APBA and amino groups of chitosan. The nanoparticles were then


SKOV-3 cells, L-02 cells

MKN45 cells, GES cells


Caco-2 cells, H22 cells

modified by iRGD and encapsulated doxorubicin. iRGD enhanced the


MDA-MB231 cells

MDA-MB231 cells

tumor penetration and inhibition of prepared nanoparticles both in 3-D


U-118 MG cells

multicellular spheroids and tumor-bearing mice compared with non-


MCF-7 cells

MCF-7 cells
Target cell

A549 cells

A549 cells

HeLa cells

conjugated ones. The in vivo tumor inhibition rate of nanoparticles was


4T1 cells

81%, while that of free drug was 24% (Wang, Zhen et al., 2013).
Chitosan oligosaccharide, depolymerized from chitosan, has low
viscosity and better aqueous solubility. Therefore, amphiphiles can be
Representative studies on chitosan-based nanoparticles for delivery of chemotherapeutic agents.

Ce6, doxorubicin, P-gp shRNA

synthesized via one-step reaction of hydrophobic modification on


methotrexate, mitomycin C

chitosan oligosaccharide. Xu et al. grafted ferrocene onto chitosan oli-


gosaccharide to fabricate stimuli-responsive nanoparticles. The nano-
demethoxycurcumin

particles could rapidly release the payloads under the synergistic effect
of low pH and oxidative stimulation (Xu, Wang, Li, & Wang, 2014).
camptothecin

gemcitabine
doxorubicin

doxorubicin
doxorubicin

doxorubicin

Furthermore, Zhu and co-workers constructed a doxorubicin-loaded


paclitaxel

paclitaxel

paclitaxel
paclitaxel

curcumin
Payload

nanoparticle based on telmisartan and stearic acid-conjugated chitosan


oligosaccharide. Telmisartan introduced nanoparticles to sequentially
target angiotensin II type I receptor (AT1R) overexpressed cancer as-
hexanoyl-chitosan-PEG, chlorotoxin-conjugated iron oxide nanoparticles

magnetic mesoporous silica nanoparticles coated with alginate/chitosan


chitosan-poly(N-3-acrylamidophenylboronic acid) modified with iRGD

sociated fibroblasts (CAFs) and tumor cells. After the elimination and
glycol chitosan-N-deoxycholic acid, octreotide-PEG-deoxycholic acid

apoptosis of CAFs, the nanoparticles acquired deeper permeability in


telmisartan and stearic acid-conjugated chitosan oligosaccharide

tumor masses and the tumor cells became more vulnerable to che-
motherapeutic agents (Zhu et al., 2018). Besides, Wang et al. in-
carboxymethylhexanoyl chitosan modified with anti-EGFR

vestigated the molecular dynamics of paclitaxel-loaded salicylic acid-


chitosan oligosaccharide nanoparticles. They found that the en-
capsulation process was majorly driven by van der Waals and hydro-
trimethyl chitosan-CSKSSDYQC (CSK) peptide

phobic interactions. Moreover, the self-assembled nanoparticles loading


PAMAM dendrimers, zwitterionic chitosan
chitosan oligosaccharide-SS-stearylamine

hydrophobic drugs were proved to be highly water-soluble, and the


chitosan oligosaccharide-indomethacin
folic acid-trimethyl chitosan-paclitaxel

optimal drug loading content was 10% (w/w) (Wang, Zhang, Wei, &
Wang, 2013).
glycol chitosan-5β-cholanic acid

polyelectrolyte multilayers
glycol chitosan-SS-octylamine

Except for being encapsulated, drugs can also be covalently con-


jugated to the hydrophilic polymers, self-assembling to form nano-
EGF-conjugated chitosan

particles, which is known as polymer-drug conjugate. The idea was


PEGylated chitosan

applied in anti-tumor drugs by Ringsdorf in 1975 (Duncan, 2006). The


conjugates usually have smaller size, which facilities deeper penetra-
tion (Wicki, Witzigmann, Balasubramanian, & Huwyler, 2014). Kwon
Material

et al. conjugated doxorubicin to glycol chitosan backbone with a cis-


Table 1

aconityl spacer, which acted as a pH-sensitive linkage. The conjugates


accumulated in tumors via EPR effect and showed long retention time in

4
E. Zhang, et al. Carbohydrate Polymers 222 (2019) 115004

Fig. 3. The structures of self-assembled nanoparticle, ionic cross-linked nanoparticle and polyelectrolyte complex.

blood circulation (Hyung Park et al., 2006; Son et al., 2003). Similarly, Saravanakumar, Kim, & Kwon, 2010; Ragelle et al., 2014; Tsai, Yu,
redox-responsive drug delivery was achieved by conjugating doxor- Huang, & Lee, 2018; Yang, Fu, Wang, & He, 2009). Unlike the chemical
ubicin to chitosan oligosaccharide via disulfide linkage (Su et al., 2015). cross-linking, the process of ionically cross-linking offers simple and
Furthermore, He et al. grafted paclitaxel and folic acid onto trimethyl mild preparation conditions without the usage of toxic reagents. Fur-
chitosan backbone for both oral and intravenous delivery of paclitaxel. thermore, the physiochemical properties of nanoparticles, such as size
The conjugates could self-assemble into nanoparticles with 18.8 folds and surface charge, can be readily customized by ionic cross-linking
higher intestinal transport of paclitaxel compared to the free drug. The with the adjustment of processing parameters, which subsequently in-
enhanced anti-tumor activity of these nanoparticles compared with the fluences drug encapsulation efficiency and release profile (Gan & Wang,
free drug was also shown in H22 tumor bearing mice (He & Yin, 2017). 2007; Gan, Wang, Cochrane, & McCarron, 2005). However, the ionic
In addition, the nanoparticles can be functionalized by conjugation cross-linked chitosan nanoparticles are weak in controlling the high
of specific ingredients to polymers. Indomethacin was a chemosensi- burst release of encapsulated drugs (Boonsongrit, Mitrevej, & Mueller,
tizer for doxorubicin in cells expressing multidrug resistance-associated 2006; Gan & Wang, 2007).
protein (MRP). A doxorubicin-loading nanoparticle based on chitosan Chen and colleagues synthesized chitosan nanoparticles using TPP
oligosaccharide-indomethacin conjugate significantly enhanced the as the cross-linker in acid solution and then chemically conjugated
anti-tumor efficacy for doxorubicin, the chemosensitizing mechanism methoxy PEG (mPEG) and methotrexate to the surface of nanoparticles.
of which was shown in Fig. 4. After intravenously injected, the nano- The in vitro and in vivo results showed the prepared nanoparticles en-
particles accumulated in tumor tissue by EPR effect, and internalized hanced the targeting ability to HeLa cells overexpressing folate re-
into tumor cells via endocytic uptake. Furthermore, intravenous injec- ceptors, approximately twice higher than folic acid-modified mPEG-
tion in rats exhibited much lower clearance of doxorubicin from na- chitosan nanoparticles (Chen et al., 2014). Similarly, Zhang et al.
noparticles than the free drug (Lee, Termsarasab et al., 2017). conjugated both mitomycin C and methotrexate to mPEG-chitosan na-
noparticles. Methotrexate not only enhanced the cellular uptake of
4.1.2. Ionic cross-linked nanoparticles nanoparticles by folic acid receptor-mediated endocytosis, but also
By exploiting the cationic property of chitosan, ionic cross-linked exerted synergistic anti-tumor efficacies together with mitomycin C
chitosan nanoparticles are fabricated through electrostatic interaction, against H22 tumors. More importantly, the nanoparticles induced no
in which the amino groups on the backbone interact with polyanionic body weight loss of mice compared with an obvious weight loss caused
cross-linking agents such as tripolyphosphate (TPP), CaCl2, Na2SO4 and by treatment of mitomycin C and methotrexate (Jia et al., 2014). Fur-
so on (Fig. 3). This method was firstly reported by Bodmeier et al. in thermore, other ligands, such as folic acid, herceptin antibody and
1989 (Bodmeier, Oh, & Pramar, 1989), and has been widely utilized in glycyrrhetinic acid, were also reported to improve tumor targeting of
preparation of drug delivery systems encapsulating proteins, peptides, ionic cross-linked chitosan nanoparticles (Arya, Vandana, Acharya, &
gene materials and low molecular drugs (Katas & Alpar, 2006; Park, Sahoo, 2011; Tian et al., 2010; Yang, Gao, & Kjems, 2014).
Additionally, Mukhtar et al. used water soluble chitosan and TPP to
fabricate nanoparticles encapsulating epigallocatechin-3-gallate
(EGCG), a polyphenol from green tea, as an oral drug delivery system.
The release of EGCG from nanoparticles was slow in stimulated gastric
juice but fast in intestinal fluid, which ensured the stability of nano-
particles in stomach. Furthermore, EGCG-loading nanoparticles ex-
hibited significant in vivo inhibiting effects against prostate and mela-
noma tumors (Khan et al., 2014; Siddiqui et al., 2014).

4.1.3. Polyelectrolyte complexes


Chitosan-based polyelectrolyte complex nanoparticles are also
formed via electrostatic interactions. After mixing chitosan with nega-
tive charged polyelectrolytes in solution, the polymeric chains interact
with each other to form strong but reversible electrostatic networks
without using cross-linkers (Fig. 3). The whole process is also facile,
simple and avoids using toxic reagents. Various polyanions have been
reported to form polyelectrolyte complex nanoparticles with chitosan,
including alginate, carrageenan, pectin, poly(acrylic acid) and so on
(Hamman, 2010). Stokke et al. investigated the factors that influence
Fig. 4. The chemosensitizing mechanism of doxorubicin-loaded chitosan oli- the physicochemical properties of polyelectrolyte complexes formed by
gosaccharide-indomethacin nanoparticles for reversing multidrug resistant chitosan and alginate. The net charge ratio and Mw of polymers were
cancer. Reproduced with permission from (Lee, Termsarasab et al., 2017). the most essential parameters that affected size, zeta potential and pH
Copyright 2017, Elsevier. Abbreviations: DOX, doxorubicin; IDM, in- of prepared complexes. Additionally, the speed and diameter of the
domethacin; CI, chitosan oligosaccharide-indomethacin conjugate. dispersing element of the homogenizer, and the mixing order might also

5
E. Zhang, et al. Carbohydrate Polymers 222 (2019) 115004

cause size change of complexes (Sæther, Holme, Maurstad, Smidsrød, &

(Jadidi-Niaragh et al.,
(Lee, Ku et al., 2017)

(Zhang et al., 2015)

(Zhang et al., 2016)


Stokke, 2008).

(Noh et al., 2011)

(Han et al., 2015)


(Wei et al., 2013)
(Lee et al., 2015)
A recent study reported the preparation of magnetic mesoporous

(Li et al., 2015)


silica nanoparticles coated with alginate/chitosan polyelectrolyte mul-

Reference
tilayers for co-delivery of P-gp shRNA, doxorubicin and photosensitizer

2017)
chlorin e6 (Ce6). The polyelectrolyte multilayers endowed this multi-
functional system with pH-sensitive property, enhanced shRNA binding
affinity and better biocompatibility. The in vivo animal studies showed

in vitro and in vivo

in vitro and in vivo

in vitro and in vivo

in vitro and in vivo

in vitro and in vivo

in vitro and in vivo

in vitro and in vivo


Biological status
that the nanoparticles exerted high therapeutic efficacy by combined
chemotherapy, photodynamic and gene therapy. Meanwhile, the Fe3O4-
Au core of the nanoparticle enabled dual-modal MR and CT imaging of

in vitro
in vivo
tumors (Yang et al., 2017). Furthermore, Yeo and co-workers mixed
polyamidoamine (PAMAM) dendrimers with zwitterionic chitosan
(ZWC) and obtained stable electrostatic complexes at pH 7.4. Interest-

combined therapy by siRNA and chemotherapeutics

suppress the expression of CD73 and potentiate the

optimize the binding affinity of siRNA and carriers


ingly, the coating of ZWC on the surface of PAMAM dendrimers reduced
its cytotoxicity against normal cells caused by high surface charge, but
the complex disintegrated at acid pH due to the charge change of ZWC.

activate mitochondria apoptosis pathway


This pH-dependent charge conversion made it potential as nanocarrier

surface charge reversible nanoparticles


delivering drugs to tumors in acid environment (Liu & Yeo, 2013).
Moreover, another study demonstrated that the coating of chitosan on

silence the expression of Mcl-1

overcome multidrug resistance


effect of dendritic cell vaccine
hyaluronic acid-paclitaxel nanoparticles could protect the conjugates

anti-angiogenesis, apoptosis
from degradation in acid environment of stomach and intestine, but
fairly fast release was shown at pH 7.4, which enhanced the bioavail-

Anti-tumor strategy
ability of orally administrated paclitaxel (Li et al., 2013).

anti-angiogenesis
4.2. Chitosan-based systems as carriers of therapeutic genes

Gene-based therapy has become an important part in the develop-


ment of oncotherapy, which is accomplished by delivering exogenous
nucleic acids (DNA, siRNA, etc.) capable of modulating gene expression

QGY-7703 cells, HeLa cells


Caco-2 cells, Lewis lung
into tumor cells. As the therapeutic nucleic acids can be easily degraded
by endonucleases in serum and the extracellular matrix, development of

HepG2/ADM cells
effective delivery vectors is the key issue for successful gene therapy
(Yin et al., 2014). Chitosan has gained large attention for delivery gene carcinoma cells
Target cell

HeLa cells

HeLa cells
PC-3 cells

PC-3 cells

products due to its non-toxicity, biodegradability, low immunogenicity


4T1 cells
KB cells

as well as the cationic property. The negatively charged DNA and siRNA
can bind to protonated amines on chitosan backbone via electrostatic
Representative studies on chitosan-based nanoparticles for delivery of chemotherapeutic genes.

interaction, which protect them from degradation by nucleases


paclitaxel, TERT siRNA

(Jayakumar et al., 2010; Ragelle et al., 2013). However, the transfec-


VEGF siRNA, Bcl-2

RAPSI nanoflower

tion efficiency of native chitosan is relatively low due to in vivo in-


stability and insufficient cellular release. Therefore, chemical mod-
Bcl-2 siRNA,
Mcl-1 siRNA

VEGF siRNA

VEGF siRNA
CD73 siRNA

P-gp siRNA,
doxorubicin
lonidamine

ifications or incorporation with functional materials are in need for


Payload

(siRNA)

developing powerful delivery systems (Ragelle et al., 2013). In recent


siRNA

years, numerous studies have reported various chitosan-based delivery


systems for delivering different DNA and siRNA with excellent anti-
urocanic acid-modified galactosylated trimethyl chitosan, poly(allylamine

tumor efficacies preclinically.


chitosan-PEI-lonidamine/triphenylphosphine, poly(acrylic acid)-PEG-folic

4.2.1. Nanoparticles based on chitosan derivatives


N-((2-hydroxy-3-trimethylammonium) propyl) chitosan chloride

Kwon et al. synthesized self-crosslinked nanoparticles based on


thiolated glycol chitosan and poly-siRNA, which was polymerized via
reducible disulfide linkages. Chitosan and poly-siRNA formed stable
complexes through both electrostatic interaction and disulfide cross-
linking. This system achieved successful in vivo delivery of Bcl-2 and
N-succinyl chitosan-poly-L-lysine-palmitic acid

vascular endothelial growth factor (VEGF) siRNA with desirable tumor


specificity and transfection efficiency (Lee et al., 2012, 2015; Yoon
hydrochloride)-citraconic anhydride
chitosan-polymer conjugates/complexes

et al., 2014). Furthermore, Kwon and colleagues prepared a polymeric


siRNA nanoflower by a novel designed rolling circle transcription (RCT)
method, which was then complexed with thiolated glycol chitosan and
α-tocopherol oligochitosan

verified for its efficacy in PC-3 xenograft tumors. The complexes


thiolated glycol chitosan

thiolated glycol chitosan

showed desirable particle stability when confronted with polyanion


Chitosan derivatives

competition or nuclease and also excellent anti-tumor activity with


chitosan-agmatine
chitosan-lactate

tumor inhibition rate of 75% at dose of 1.5 mg/kg. (Lee, Ku et al.,


2017).
acid
Material

Trimethylated chitosan, a water soluble chitosan derivative, has


Table 2

enhanced complexing ability between chitosan and anionic nucleic acid


(Gao, Wang et al., 2014). Yin et al. prepared a series of galactosylated

6
E. Zhang, et al. Carbohydrate Polymers 222 (2019) 115004

Fig. 5. The structure of chitosan-PEI nanoparticle co-delivering lonidamine and Bcl-2 siRNA (A). The process of hierarchically targeting and mechanism of mi-
tochondrial apoptosis pathway induced by Bcl-2 siRNA and lonidamine (B). Reproduced with permission from (Zhang et al., 2015). Copyright 2015, Elsevier.
Abbreviations: PAA, poly(acrylic acid); PPF, poly(acrylic acid)-polyethylene glycol-folic acid; LND, lonidamine; mPTP, mitochondrial permeability transition pore;
RISC, RNA-induced silencing complex.

trimethyl chitosan-cysteine/siRNA complexes to study the effect of 4.2.2. Nanoparticles based on chitosan-polymer conjugates/complexes
siRNA binding affinity on anti-tumor efficacy. The strong binding The conjugation or addition of functional polymers in chitosan
strength provided excellent stability of complexes in serum but in- formulations can cover the shortage of chitosan and achieve more ef-
efficient release of siRNA inside the cells, while the weak binding ficient transfection. Poly(ethylene imine) (PEI) is one of the most stu-
strength came to the opposite results. The authors suggested that tai- died amine-rich polymeric materials for nucleic acid delivery. Due to
loring the binding strength between carriers and siRNA was important the abundant amino groups on its chains, PEI has high charge intensity
for improving transfection efficiency and anti-tumor activity (Han, at sponge effect. Nevertheless, PEI is also known to be toxic against
Tang, & Yin, 2013). normal cells due to the large positive charge. The incorporation of PEI
Furthermore, Ren et al. prepared folic acid-modified hydroxypropyl with chitosan nanoparticles is often carried out via either chemical
chitosan nanoparticles delivering antisense oligodeoxynucleotides for modification or addition as an excipient. PEI improved the endosomal
overcoming tumor drug resistance. The nanoparticles selectively accu- escape capacity of chitosan nanoparticles and the subsequent trans-
mulated in folic acid receptor-high expressing tumor cells and inhibited fection efficiency, and also, the cytotoxicity is reduced compared with
multi drug resistance (MDR) 1 gene and P-glycoprotein (P-gp) levels native PEI nanoparticles (Buschmann et al., 2013; Huh et al., 2010).
(Wang, Tao, Zhang, Wei, & Ren, 2010). Moreover, N-(2-hydroxy-3-tri- Jiang et al. designed a lonidamine-conjugated chitosan-g-PEI nano-
methylammonium)propyl chitosan chloride, a partially quaternized particle for Bcl-2 siRNA delivery, hierarchically targeting to tumor cells
chitosan, was utilized for oral delivery of mouse telomerase reverse and mitochondria. The structure and targeting mechanism were dis-
transcriptase (mTERT) siRNA and paclitaxel simultaneously. This played in Fig. 5. After folic acid-mediated internalization into target
chitosan derivative enhanced intestinal absorption and cellular uptake, cells, lonidamine was delivering into mitochondria by triphenylpho-
which resulted in synergistic anti-tumor effect by siRNA and paclitaxel sphine (TPP) and induced apoptosis. Meanwhile, siRNA was released
(Wei et al., 2013). into cytoplasm and suppressed anti-apoptotic Bcl-2 expression, re-
In addition, Li and co-workers prepared pH-sensitive charge-con- sulting in significant anti-tumor activity via mitochondrial signaling
versional nanocomplexes based on dimethylmaleic anhydride modified pathway synergistically with lonidamine. The proliferation rate of HeLa
chitosan-agmatine conjugates, which could condense VEGF siRNA into cells treated by the nanoparticles after 72 h was only 11.9%, which was
nanoparticles. The zeta potential of nanoparticles transformed from significantly lower than groups treated by lonidamine or siRNA (Zhang
negative charge into positive charge at acid pH, facilitating tumor et al., 2015). Similarly, cationic polymers like poly-L-arginine (PLR)
cellular uptake and subsequent VEGF mRNA knockdown (approxi- (Noh et al., 2010), poly-L-lysine (PLL) (Zhang et al., 2016) and poly-
mately 79.7%) and cell apoptosis (Li et al., 2015). guluronate (Lee et al., 2009) were used to increase gene stability and

7
E. Zhang, et al. Carbohydrate Polymers 222 (2019) 115004

transfection efficiency of chitosan nanoparticles.

(Hsiao, Chuang et al.,


(Wang et al., 2015)
(Ding et al., 2018)
Except for cationic polymers, some anionic polymers were reported

(Gao et al., 2016)


(Zhang, Lv et al.,

(Liu et al., 2018)

(Shi et al., 2017)


(Xu et al., 2012)
(Li et al., 2017)
to improve the in vivo performance of chitosan/siRNA systems. Yin et al.
incorporated anionic poly(allylamine hydrochloride)-citraconic anhy-
Reference

2017) dride (PAH-Cit) with urocanic acid-conjugated galactosylated trimethyl

2015)
chitosan for delivery of VEGF siRNA. PAH-Cit could enhanced the en-
dosomal escape capability due to its property of hydrolyzing into ca-

in vitro anti-tumor activity and in vivo


tionic poly(allylamine) in endosomes (Han, Tang, & Yin, 2015). Fur-
thermore, Babu et al. coated negatively charged polylactic acid
nanoparticles with cationic chitosan shells, co-delivering P62siRNA, β5
plasmid DNA and cisplatin for reversing cancer drug resistance. After
the release of therapeutic agents into cisplatin-resistant 2008/C13
in vitro and in vivo

in vitro and in vivo

in vitro and in vivo

in vitro and in vivo

in vitro and in vivo


ovarian cancer cells, the treatment with these nanoparticles caused P62
biocompatibility
Biological status

inhibition and restoration of β5 expression, which significantly en-


hanced the anti-tumor activity of cisplatin (Babu et al., 2014). More-
in vitro
in vitro

over, the effectiveness of chitosan/siRNA nanoparticles was also im-


in vivo

proved when incorporated with anionic poly(γ-glutamic acid) (γ-PGA).


The incorporation of γ-PGA improved the cellular uptake, intracellular
chemo-photothermal therapy, two-

unpackage, transfection efficiency compared with chitosan/siRNA na-


therapy for deep-

noparticles. In particular, chitosan/γ-PGA based siRNA nanoparticles


exhibited a knockdown efficiency of 72% after 24 h post transfection
imaging,

and 55% after 72 h, compared with chitosan/siRNA nanoparticles with


photodynamic therapy,
therapy

therapy

therapy

photothermal therapy

photothermal therapy
Anti-tumor strategy

50% after 24 h and only 30% after 72 h (Liao et al., 2010).


photon bioimaging

immunotherapy
immunotherapy
photodynamic

photodynamic
photodynamic

photodynamic
tumor-specific

seated tumors

4.3. Chitosan-based systems as carriers of other therapeutic agents

Photodynamic therapy is a rapidly growing treatment for oncology


due to its minimal toxicity compared with chemotherapy and radio-
therapy. Selective delivery of water-insoluble photosensitizers into
target tissues is one of the greatest challenges for successful therapeutic
B16 cells, bone marrow
HepG2 cells, H22 cells

derived dendritic cells


A549 cells, L02 cells

outcome (Agostinis et al., 2011; Lucky, Soo, & Zhang, 2015). Lee at al.
prepared an intelligent polymer-drug conjugate based on glycol chit-
osan, on which a pH-sensitive block, 3-diethylaminopropyl iso-
DU145 cells

Hep3B cells
MCF-7 cells
Target cell

CT26 cells
HeLa cells

PC-3 cells

thiocyanate (DEAP) group, photosensitizer Ce6 and PEG was grafted.


The conformation of conjugates could switch from self-assembled na-
Representative studies on chitosan-based nanoparticles for delivery of other chemotherapeutic agents.

noparticles at neutral pH into uncoiled molecules when exposed in acid


environment. Based on this property, the conjugates exhibited pH-in-
dependent singlet-oxygen generation, which was crucial for photo-
toxicity against tumor cells (Park et al., 2011). Furthermore, Ding et al.
prepared an amphiphilic polymer by conjugating Ce6 onto glycol
whole tumor cell lysate
heptamethine cyanine

mono-hydroxylphenyl

chitosan backbone, which subsequently self-assembled into nano-


Zinc phthalocyanine

triphenylporphyrin
indocyanine green

particles. The cytotoxicity of Ce6 was markedly attenuated but the


doxorubicin, CD

therapeutic efficacy was improved (Ding et al., 2018). Moreover, in


polyaniline

order to improve efficiency of photodynamic therapy against deep-se-


Payload

ated tumors, Gao and co-workers synthesized a novel nanosystems


IL-12
Ce6

consisting of Zinc phthalocyanine-loaded nanoparticles coated with c


(RGDyK)-conjugated N-succinyl-N-octyl chitosan. A significant im-
Zinc phthalocyanine nanoparticle coated with c(RGDyK)-N-succinyl-

DMA-chitosan oligosaccharide-PEG as the shell, mesoporous silica

provement of therapeutic efficacy by this nanosystem was shown


mono-hydroxylphenyl triphenylporphyrin-chitosan carbon dots

compared with traditional photodynamic therapy. The authors also


introduced a sequential treatment of nanosystem followed by Doxil,
which could simultaneously enhance the efficacy with a tumor inhibi-
coated gold nanorod modified with peptide RLA

tion rate of 79% compared with 56% of Doxil alone, and also reduced
folic acid/heptamethine cyanine-modified chitosan

the cardiotoxicity of Doxil (Gao et al., 2016).


As an extension of photodynamic therapy, photothermal therapy
has also attracted large attention for precise cancer treatment. Liu et al.
utilized chitosan oligosaccharide-PEG-2,3-dimethylmaleic anhydride
PEG-chitosan coated CD nanogels

(DMA) copolymer as a shell protecting mesoporous silica coated gold


nanorods for delivery of indocyanine green. The composition and in
mannose- alginate, chitosan

vivo working process of this nanosystem was shown in Fig. 6. The


N- polyaniline-chitosan

chitosan-based outer shell could prevent the nanoparticles from serum


N-octyl chitosan
glycol chitosan-Ce6

chitosan glutamate

absorption and prolonged the blood circulation time. The amide bonds
between chitosan and DMA broke down in acidic microenvironment,
resulting in the decomposition of the shell. This composite nanosystem
Material

achieved successful photodynamic and photothermal therapy for breast


Table 3

cancer at the same time with minimal side effects (Liu et al., 2018).
Furthermore, Wang and colleagues prepared PEG-chitosan coated

8
E. Zhang, et al. Carbohydrate Polymers 222 (2019) 115004

Fig. 6. The preparation and in vivo working process of a multi-


functional nanosystem for simultaneous photodynamic and pho-
tothermal therapy against breast cancer. Reproduced with per-
mission from (Liu et al., 2018). Copyright 2018, Elsevier.
Abbreviations: AuNR, gold nanorods; MSN, ; APTES, 3-amino-
propyltriethoxysilane; ICG, indocyanine green; β-CD, β-cyclodex-
trin; PTT, photothermal therapy; PDT, photodynamic therapy (For
interpretation of the references to colour in this figure legend, the
reader is referred to the web version of this article).

carbon dots (CD) nanogels loading doxorubicin for pH and NIR light- taken up by dendritic cells and increase serum IFN-γ and IL-4. Fur-
sensitive drug release, bioimaging, and chemo-photothermal therapy. thermore, this novel vaccine significantly delayed the tumor growth
The hydroxyl and amine groups of chitosan help form hydrogen bonds partially due to cytotoxic T lymphocytes response (Shi et al., 2017).
with hydroxyl and carboxyl groups of CDs as well as ether oxygens of
PEG. Besides, the abundant amine groups on chitosan could modulate
5. Challenges of translation from laboratory to clinic
the drug release by pH-sensitive swelling/deswelling effect. This novel
nanosystem provided high anti-tumor activity against DU145 prostate
Till now, there were no chitosan-based nanoparticles approved for
cancer cells due to the synergistic effect of chemo-photothermal
treatment of cancers in clinic or being tested clinical trials. The clinical
therapy (Wang et al., 2015). Moreover, Lu et al. designed a chitosan-
translation of anti-tumor chitosan nanoparticles is confronted with
coated CuS nanoparticles incorporated with immunoadjuvants, oligo-
many challenges. First of all, there are only a few studies testing the in
deoxynucleotides containing cytosine-guanine (CpG) motifs, which
vivo safety of chitosan, especially chitosan administrated by intravenous
exerted anti-tumor activity through both photothermal and im-
injection, because most preclinical anti-tumor chitosan-based nano-
munotherapy. The nanoparticles were disintegrated after laser excita-
particles as described in this review were designed as injections.
tion, allowing the system reassembled into chitosan-CpG complexes,
Additionally, the chemical versatility of chitosan makes it more difficult
subsequently activated host antitumor immunity. The combined
to carried out, because each modification produced a new chemical
therapy showed more effective than immunotherapy or photothermal
entity which should be assessed individually. However, some dis-
therapy alone, resulting in synergistic effects against both primary-
advantages of chitosan, such as the poor water solubility and inefficient
treated and distant-untreated tumors (Guo et al., 2014).
endosomal escaping ability, compel it to make structural modification.
Chitosan and its derivatives also attract large attention as drug
Furthermore, chitosan-based nanoparticles shared some common
carriers for cancer immunotherapy. Interleukin-12 (IL-12) is a cytokine
problems with other nano-drug delivery systems. For example, the
with immunomodulatory and angiogenesis activities, and it has been
targeting mechanisms which are considered as the rationale of design
widely investigated as either an anti-tumor agent or a vaccine adjuvant.
and development of anti-tumor nanomedicine, like EPR effect and ac-
However, the systematic toxicity of IL-12 seriously hindered its clinical
tive targeting, are often being questioned (Danhier, 2016; Mcneil, 2016;
applications (‘Mac’Cheever, 2008; Del Vecchio et al., 2007). Wang et al.
Wilhelm, Tavares, & Chan, 2016, 2016b). It is still uncertain whether
incorporated IL-12 with chitosan glutamate nanoparticles using TPP as
the targeting mechanisms of nanoparticles displayed in rodents works
cross-linker. The intravenously administrated nanoparticles sig-
in humans due to the heterogeneity of clinical tumors. Therefore, taking
nificantly inhibited the growth of liver metastasis tumors of colorectal
a deep insight into the correlation of patient tumors and nanomedicine
cancer and reduced the toxicity caused by IL-12. Furthermore, in-
behavior is necessary for further clinical translation.
filtration of microphages and T cells was induced during the im-
In addition, the large-scale production is another problem for clin-
munotherapy, which might be related to the tumor inhibition (Xu et al.,
ical translation of chitosan-based nanoparticles. Chitosan, composed of
2012). Besides, in order to enhance the immunotherapeutic efficacy of
a broad range of poly-N-acetyl-glucosamine, varies in Mw and DD,
tumor cell lysates (TCL), Shi et al. encapsulated TCL into nanoparticles
which depends on the sources, extraction and preparation processes,
based on mannose-modified alginate and chitosan, targeting specific
and its derivatives change from degrees of substitutions. Improved
dendritic cells. The prepared nanoparticles vaccine could be readily
processing and characterization in production are in need.

9
E. Zhang, et al. Carbohydrate Polymers 222 (2019) 115004

Furthermore, compared to traditional chemotherapeutic agents, the 277, 142–153.


reproducibility and quality control of nanoparticles are far more com- Chen, J., Huang, L., Lai, H., Lu, C., Fang, M., Zhang, Q., et al. (2014). Methotrexate-
loaded PEGylated chitosan nanoparticles: Synthesis, characterization, and in vitro
plicated. How to retain the batch-to-batch consistency is a key problem and in vivo antitumoral activity. Molecular Pharmaceutics, 11, 2213–2223.
for large-scale production. Besides, the high cost of large-scale pro- Danhier, F. (2016). To exploit the tumor microenvironment: since the EPR effect fails in
duction discouraged pharmaceutical companies from clinical transla- the clinic, what is the future of nanomedicine? Journal of Controlled Release, 244,
108–121.
tion of nanomedicine. Del Vecchio, M., Bajetta, E., Canova, S., Lotze, M. T., Wesa, A., Parmiani, G., et al. (2007).
Interleukin-12: Biological properties and clinical application. Clinical Cancer
6. Conclusion and perspectives Research, 13, 4677–4685.
Ding, Y., Li, S., Liang, L., Huang, Q., Yuwen, L., Yang, W., et al. (2018). Highly bio-
compatible chlorin e6-loaded chitosan nanoparticles for improved photodynamic
Chitosan-based nanoparticles present many advantages for anti- cancer therapy. ACS Applied Materials & Interfaces, 10, 9980–9987.
tumor drug delivery. Various anti-tumor agents, such as hydrophilic or Duncan, R. (2006). Polymer conjugates as anticancer nanomedicines. Nature Reviews
Cancer, 6, 688–701.
hydrophobic chemotherapeutic drugs, nucleic acids, photosensitizers
Fernandes, J. C., Qiu, X., Winnik, F. M., Benderdour, M., Zhang, X., Dai, K., et al. (2012).
and cytokines, can be incorporated into chitosan-based nanoparticles Low molecular weight chitosan conjugated with folate for siRNA delivery in vitro:
and delivered to specific tumor cells via EPR effect and ligand-receptor Optimization studies. International Journal of Nanomedicine, 7, 5833–5845.
interaction. Furthermore, the cationic property of chitosan facilitates Gan, Q., & Wang, T. (2007). Chitosan nanoparticle as protein delivery carrier-systematic
examination of fabrication conditions for efficient loading and release. Colloids and
endosomal escape of nanoparticles and the efficient release of nucleic Surfaces B, Biointerfaces, 59, 24–34.
acids into cytoplasm. Numerous studies demonstrated that chitosan- Gan, Q., Wang, T., Cochrane, C., & McCarron, P. (2005). Modulation of surface charge,
based nanoparticles not only caused obvious side effects on experi- particle size and morphological properties of chitosan-TPP nanoparticles intended for
gene delivery. Colloids and Surfaces B, Biointerfaces, 44, 65–73.
mental animals, but also alleviated the toxicity of chemotherapeutic Gao, C., Liu, T., Dang, Y., Yu, Z., Wang, W., Guo, J., et al. (2014). pH/redox responsive
agents or other ingredients like PEI. In a number of cases, chitosan- core cross-linked nanoparticles from thiolated carboxymethyl chitosan for in vitro
based nanoparticles exhibited exciting in vivo anti-tumor efficacy and release study of methotrexate. Carbohydrate Polymers, 111, 964–970.
Gao, W., Wang, Z., Lv, L., Yin, D., Chen, D., Han, Z., et al. (2016). Photodynamic therapy
favorable targeting ability, which indicated there is tremendous scope induced enhancement of tumor vasculature permeability using an upconversion na-
in clinical application. noconstruct for improved intratumoral nanoparticle delivery in deep tissues.
Overall, numerous studies presented exciting anti-tumor efficacy of Theranostics, 6, 1131–1144.
Gao, Y., Wang, Z.-Y., Zhang, J., Zhang, Y., Huo, H., Wang, T., et al. (2014). RVG-peptide-
chitosan-based nanoparticles in recent years. Chitosan and its deriva- linked trimethylated chitosan for delivery of siRNA to the brain. Biomacromolecules,
tives are capable of targeting delivery of various anti-tumor agents, 15, 1010–1018.
which can be applied to a variety of treatment for oncotherapy. The Garcia-Fuentes, M., & Alonso, M. J. (2012). Chitosan-based drug nanocarriers: where do
we stand? Journal of Controlled Release, 161, 496–504.
future studies should be focus on not only development of more effi-
Gjoseva, S., Geskovski, N., Sazdovska, S. D., Popeski-Dimovski, R., Petruševski, G.,
cient and simple chitosan-based nanoparticles for oncotherapy, but also Mladenovska, K., et al. (2018). Design and biological response of doxycycline loaded
their safety after intravenously administrated as well as targeting me- chitosan microparticles for periodontal disease treatment. Carbohydrate Polymers,
chanisms by using more clinical-relevant tumor models. 186, 260–272.
Guo, L., Yan, D. D., Yang, D., Li, Y., Wang, X., Zalewski, O., et al. (2014). Combinatorial
photothermal and immuno cancer therapy using chitosan-coated hollow copper
Acknowledgements sulfide nanoparticles. ACS Nano, 8, 5670–5681.
Hamman, J. H. (2010). Chitosan based polyelectrolyte complexes as potential carrier
materials in drug delivery systems. Marine Drugs, 8, 1305–1322.
This work is supported by National Key R&D Program of China Han, L., Tang, C., & Yin, C. (2013). Effect of binding affinity for siRNA on the in vivo
(2018YFC0311305); Special projects of Foshan science and technology antitumor efficacy of polyplexes. Biomaterials, 34, 5317–5327.
innovation team (2017IT100054). Han, L., Tang, C., & Yin, C. (2015). Enhanced antitumor efficacies of multifunctional
nanocomplexes through knocking down the barriers for siRNA delivery. Biomaterials,
44, 111–121.
References He, P., Davis, S. S., & Illum, L. (1998). In vitro evaluation of the mucoadhesive properties
of chitosan microspheres. International Journal of Pharmaceutics, 166, 75–88.
He, R., & Yin, C. (2017). Trimethyl chitosan based conjugates for oral and intravenous
Agostinis, P., Berg, K., Cengel, K. A., Foster, T. H., Girotti, A. W., Gollnick, S. O., et al.
delivery of paclitaxel. Acta Biomaterialia, 53, 355–366.
(2011). Photodynamic therapy of cancer: An update. CA: A Cancer Journal for
Hoppe-Seyler, F. (1894). Ueber chitin und cellulose. Berichte Der Deutschen Chemischen
Clinicians, 61, 250–281.
Gesellschaft, 27, 3329–3331.
Alameh, M., Lavertu, M., Tran-Khanh, N., Chang, C.-Y., Lesage, F., Bail, M., et al. (2018).
Hsiao, C.-W., Chuang, E.-Y., Chen, H.-L., Wan, D., Korupalli, C., Liao, Z.-X., et al. (2015).
siRNA delivery with chitosan: Influence of chitosan molecular weight, degree of
Photothermal tumor ablation in mice with repeated therapy sessions using NIR-ab-
deacetylation, and amine to phosphate ratio on in vitro silencing efficiency, hemo-
sorbing micellar hydrogels formed in situ. Biomaterials, 56, 26–35.
compatibility, biodistribution, and in vivo efficacy. Biomacromolecules, 19, 112–131.
Hsiao, M.-H., Mu, Q., Stephen, Z. R., Fang, C., & Zhang, M. (2015). Hexanoyl-chitosan-
American Society for Testing and Materials (2011). Standard guide for characterization and
PEG copolymer coated iron oxide nanoparticles for hydrophobic drug delivery. ACS
testing of chitosan salts as starting materials intended for use in biomedical and tissue-
Macro Letters, 4, 403–407.
engineered medical product applications. 1–8 America.
Hu, Y. W., Du, Y. Z., Liu, N., Liu, X., Meng, T. T., Cheng, B. L., et al. (2015). Selective
Arya, G., Vandana, M., Acharya, S., & Sahoo, S. K. (2011). Enhanced antiproliferative
redox-responsive drug release in tumor cells mediated by chitosan based glycolipid-
activity of Herceptin (HER2)-conjugated gemcitabine-loaded chitosan nanoparticle in
like nanocarrier. Journal of Controlled Release, 206, 91–100.
pancreatic cancer therapy. Nanomedicine Nanotechnology Biology and Medicine, 7,
Huang, M., Fong, C.-W., Khor, E., & Lim, L.-Y. (2005). Transfection efficiency of chitosan
859–870.
vectors: Effect of polymer molecular weight and degree of deacetylation. Journal of
Babu, A., Wang, Q., Muralidharan, R., Shanker, M., Munshi, A., & Ramesh, R. (2014).
Controlled Release, 106, 391–406.
Chitosan coated polylactic acid nanoparticle-mediated combinatorial delivery of
Huang, M., Khor, E., & Lim, L. Y. (2004). Uptake and cytotoxicity of chitosan molecules
cisplatin and siRNA/Plasmid DNA chemosensitizes cisplatin-resistant human ovarian
and nanoparticles: Effects of molecular weight and degree of deacetylation.
cancer cells. Molecular Pharmaceutics, 11, 2720–2733.
Pharmaceutical Research, 21, 344–353.
Baldrick, P. (2010). The safety of chitosan as a pharmaceutical excipient. Regulatory
Huang, W.-T., Larsson, M., Wang, Y.-J., Chiou, S.-H., Lin, H.-Y., & Liu, D.-M. (2015).
Toxicology and Pharmacology, 56, 290–299.
Demethoxycurcumin-carrying chitosan-antibody core-shell nanoparticles with mul-
Bei, Y. Y., Yuan, Z. Q., Zhang, L., Zhou, X. F., Chen, W. L., Xia, P., et al. (2014). Novel self-
titherapeutic efficacy toward malignant A549 lung tumor: From in vitro character-
assembled micelles based on palmitoyl-trimethyl-chitosan for efficient delivery of
ization to in vivo evaluation. Molecular Pharmaceutics, 12, 1242–1249.
harmine to liver cancer. Expert Opinion on Drug Delivery, 11, 843–854.
Huh, M. S., Lee, S.-Y., Park, S., Lee, S., Chung, H., Lee, S., et al. (2010). Tumor-homing
Bodmeier, R., Oh, K. H., & Pramar, Y. (1989). Preparation and evaluation of drug-con-
glycol chitosan/polyethylenimine nanoparticles for the systemic delivery of siRNA in
taining chitosan beads. Drug Development Communications, 15, 1475–1494.
tumor-bearing mice. Journal of Controlled Release, 144, 134–143.
Boonsongrit, Y., Mitrevej, A., & Mueller, B. W. (2006). Chitosan drug binding by ionic
Huo, M., Liu, Y., Wang, L., Yin, T., Qin, C., Xiao, Y., et al. (2016). Redox-sensitive micelles
interaction. European Journal of Pharmaceutics and Biopharmaceutics, 62, 267–274.
based on O,N-hydroxyethyl chitosan–octylamine conjugates for triggered in-
Buschmann, M. D., Merzouki, A., Lavertu, M., Thibault, M., Jean, M., & Darras, V. (2013).
tracellular delivery of paclitaxel. Molecular Pharmaceutics, 13, 1750–1762.
Chitosans for delivery of nucleic acids. Advanced Drug Delivery Reviews, 65,
Huo, M., Zou, A., Yao, C., Zhang, Y., Zhou, J., Wang, J., et al. (2012). Somatostatin
1234–1270.
receptor-mediated tumor-targeting drug delivery using octreotide-PEG-deoxycholic
Chen, G., Svirskis, D., Lu, W., Ying, M., Huang, Y., & Wen, J. (2018). N-trimethyl chitosan
acid conjugate-modified N-deoxycholic acid-O, N-hydroxyethylation chitosan mi-
nanoparticles and CSKSSDYQC peptide: N-trimethyl chitosan conjugates enhance the
celles. Biomaterials, 33, 6393–6407.
oral bioavailability of gemcitabine to treat breast cancer. Journal of Controlled Release,
Hwang, H. Y., Kim, I. S., Kwon, I. C., & Kim, Y. H. (2008). Tumor targetability and

10
E. Zhang, et al. Carbohydrate Polymers 222 (2019) 115004

antitumor effect of docetaxel-loaded hydrophobically modified glycol chitosan na- multifunctional nanoplatform for NIR-mediated multiple enhanced photodynamic
noparticles. Journal of Controlled Release, 128, 23–31. and photothermal tumor therapy. Biomaterials, 157, 107–124.
Hyung Park, J., Kwon, S., Lee, M., Chung, H., Kim, J. H., Kim, Y. S., et al. (2006). Self- Liu, K. C., & Yeo, Y. (2013). Zwitterionic chitosan-polyamidoamine dendrimer complex
assembled nanoparticles based on glycol chitosan bearing hydrophobic moieties as nanoparticles as a pH-sensitive drug carrier. Molecular Pharmaceutics, 10, 1695–1704.
carriers for doxorubicin: In vivo biodistribution and anti-tumor activity. Biomaterials, Liu, X., Howard, K. A., Dong, M., Andersen, M. O., Rahbek, U. L., Johnsen, M. G., et al.
27, 119–126. (2007). The influence of polymeric properties on chitosan/siRNA nanoparticle for-
Jadidi-Niaragh, F., Atyabi, F., Rastegari, A., Kheshtchin, N., Arab, S., Hassannia, H., et al. mulation and gene silencing. Biomaterials, 28, 1280–1288.
(2017). CD73 specific siRNA loaded chitosan lactate nanoparticles potentiate the Lucky, S. S., Soo, K. C., & Zhang, Y. (2015). Nanoparticles in photodynamic therapy.
antitumor effect of a dendritic cell vaccine in 4T1 breast cancer bearing mice. Journal Chemical Reviews, 115, 1990–2042.
of Controlled Release, 246, 46–59. ‘Mac’Cheever, M. A. (2008). Twelve immunotherapy drugs that could cure cancers.
Jayakumar, R., Chennazhi, K. P., Muzzarelli, R. A. A., Tamura, H., Nair, S. V., & Immunological Reviews, 222, 357–368.
Selvamurugan, N. (2010). Chitosan conjugated DNA nanoparticles in gene therapy. Malmo, J., Sørgård, H., Vårum, K. M., & Strand, S. P. (2012). siRNA delivery with chit-
Carbohydrate Polymers, 79, 1–8. osan nanoparticles: Molecular properties favoring efficient gene silencing. Journal of
Jia, M., Li, Y., Yang, X., Huang, Y., Wu, H., Huang, Y., et al. (2014). Development of both Controlled Release, 158, 261–268.
methotrexate and mitomycin C loaded PEGylated chitosan nanoparticles for targeted Mao, S., Sun, W., & Kissel, T. (2010). Chitosan-based formulations for delivery of DNA
drug codelivery and synergistic anticancer effect. ACS Applied Materials & Interfaces, and siRNA. Advanced Drug Delivery Reviews, 62, 12–27.
6, 11413–11423. Mcneil, S. E. (2016). Evaluation of nanomedicines: Stick to the basics. Nature Reviews
Katas, H., & Alpar, H. O. (2006). Development and characterisation of chitosan nano- Materials, 1, 16073.
particles for siRNA delivery. Journal of Controlled Release, 115, 216–225. Min, K. H., Park, K., Kim, Y. S., Bae, S. M., Lee, S., Jo, H. G., et al. (2008).
Kean, T., & Thanou, M. (2010). Biodegradation, biodistribution and toxicity of chitosan. Hydrophobically modified glycol chitosan nanoparticles-encapsulated camptothecin
Advanced Drug Delivery Reviews, 62, 3–11. enhance the drug stability and tumor targeting in cancer therapy. Journal of
Key, J., Dhawan, D., Cooper, C. L., Knapp, D. W., Kim, K., Kwon, I. C., et al. (2016). Controlled Release, 127, 208–218.
Multicomponent, peptide-targeted glycol chitosan nanoparticles containing ferri- Na, J. H., Koo, H., Lee, S., Han, S. J., Lee, K. E., Kim, S., et al. (2016). Precise targeting of
magnetic iron oxide nanocubes for bladder cancer multimodal imaging. International liver tumor using glycol chitosan nanoparticles: Mechanisms, key factors, and their
Journal of Nanomedicine, 11, 4141–4155. implications. Molecular Pharmaceutics, 13, 3700–3711.
Khan, N., Bharali, D. J., Adhami, V. M., Siddiqui, I. A., Cui, H., Shabana, S. M., et al. Na, J. H., Lee, S.-Y., Lee, S., Koo, H., Min, K. H., Jeong, S. Y., et al. (2012). Effect of the
(2014). Oral administration of naturally occurring chitosan-based nanoformulated stability and deformability of self-assembled glycol chitosan nanoparticles on tumor-
green tea polyphenol EGCG effectively inhibits prostate cancer cell growth in a xe- targeting efficiency. Journal of Controlled Release, 163, 2–9.
nograft model. Carcinogenesis, 35, 415–423. Narayanan, D., Jayakumar, R., & Chennazhi, K. P. (2014). Versatile carboxymethyl chitin
Kiang, T., Wen, J., Lim, H. W., & Leong, K. W. (2004). The effect of the degree of chitosan and chitosan nanomaterials: A review. Wiley Interdisciplinary Reviews-Nanomedicine
deacetylation on the efficiency of gene transfection. Biomaterials, 25, 5293–5301. and Nanobiotechnology, 6, 574–598.
Kim, J.-H., Kim, Y.-S., Park, K., Lee, S., Nam, H. Y., Min, K. H., et al. (2008). Antitumor Noh, S. M., Han, S. E., Shim, G., Lee, K. E., Kim, C.-W., Han, S. S., et al. (2011).
efficacy of cisplatin-loaded glycol chitosan nanoparticles in tumor-bearing mice. Tocopheryl oligochitosan-based self assembling oligomersomes for siRNA delivery.
Journal of Controlled Release, 127, 41–49. Biomaterials, 32, 849–857.
Kim, J. K., Han, K. H., Lee, J. T., Paik, Y. H., Ahn, S. H., Lee, J. D., et al. (2006). Long-term Noh, S. M., Park, M. O., Shim, G., Han, S. E., Lee, H. Y., Huh, J. H., et al. (2010). Pegylated
clinical outcome of phase IIb clinical trial of percutaneous injection with holmium- poly-l-arginine derivatives of chitosan for effective delivery of siRNA. Journal of
166/chitosan complex (Milican) for the treatment of small hepatocellular carcinoma. Controlled Release, 145, 159–164.
Clinical Cancer Research, 12, 543–548. Park, J. H., Saravanakumar, G., Kim, K., & Kwon, I. C. (2010). Targeted delivery of low
Kim, K., Kwon, S., Park, J. H., Chung, H., Jeong, S. Y., Kwon, I. C., et al. (2005). molecular drugs using chitosan and its derivatives. Advanced Drug Delivery Reviews,
Physicochemical characterizations of self-assembled nanoparticles of glycol chitosan- 62, 28–41.
deoxycholic acid conjugates. Biomacromolecules, 6, 1154–1158. Park, K., Kim, J. H., Nam, Y. S., Lee, S., Nam, H. Y., Kim, K., et al. (2007). Effect of
Koo, H., Min, K. H., Lee, S. C., Park, J. H., Park, K., Jeong, S. Y., et al. (2013). Enhanced polymer molecular weight on the tumor targeting characteristics of self-assembled
drug-loading and therapeutic efficacy of hydrotropic oligomer-conjugated glycol glycol chitosan nanoparticles. Journal of Controlled Release, 122, 305–314.
chitosan nanoparticles for tumor-targeted paclitaxel delivery. Journal of Controlled Park, S. Y., Baik, H. J., Oh, Y. T., Oh, K. T., Youn, Y. S., & Lee, E. S. (2011). A smart
Release, 172, 823–831. polysaccharide/drug conjugate for photodynamic therapy. Angewandte Chemie
Köping-Höggård, M., Vårum, K. M., Issa, M., Danielsen, S., Christensen, B. E., Stokke, B. International Edition, 50, 1644–1647.
T., et al. (2004). Improved chitosan-mediated gene delivery based on easily dis- Ragelle, H., Riva, R., Vandermeulen, G., Naeye, B., Pourcelle, V., Le Duff, C. S., et al.
sociated chitosan polyplexes of highly defined chitosan oligomers. Gene Therapy, 11, (2014). Chitosan nanoparticles for siRNA delivery: Optimizing formulation to in-
1441. crease stability and efficiency. Journal of Controlled Release, 176, 54–63.
Lee, D. W., Yun, K.-S., Ban, H.-S., Choe, W., Lee, S. K., & Lee, K. Y. (2009). Preparation Ragelle, H., Vandermeulen, G., & Préat, V. (2013). Chitosan-based siRNA delivery sys-
and characterization of chitosan/polyguluronate nanoparticles for siRNA delivery. tems. Journal of Controlled Release, 172, 207–218.
Journal of Controlled Release, 139, 146–152. Ravindranathan, S., Koppolu, B. P., Smith, S. G., & Zaharoff, D. A. (2016). Effect of
Lee, J.-Y., Termsarasab, U., Lee, M. Y., Kim, D.-H., Lee, S. Y., Kim, J. S., et al. (2017). chitosan properties on immunoreactivity. Marine Drugs, 14, 91.
Chemosensitizing indomethacin-conjugated chitosan oligosaccharide nanoparticles Rouget, C. (1859). Des substances amylacées dans le tissu des animaux, spécialement les
for tumor-targeted drug delivery. Acta Biomaterialia, 57, 262–273. articulés (chitine). Comp. Rend. 48, 792–795.
Lee, J. H., Ku, S. H., Kim, M. J., Lee, S. J., Kim, H. C., Kim, K., et al. (2017). Rolling circle Rudzinski, W. E., & Aminabhavi, T. M. (2010). Chitosan as a carrier for targeted delivery
transcription-based polymeric siRNA nanoparticles for tumor-targeted delivery. of small interfering RNA. International Journal of Pharmaceutics, 399, 1–11.
Journal of Controlled Release, 263, 29–38. Sæther, H. V., Holme, H. K., Maurstad, G., Smidsrød, O., & Stokke, B. T. (2008).
Lee, S. J., Huh, M. S., Lee, S. Y., Min, S., Lee, S., Koo, H., et al. (2012). Tumor-homing Polyelectrolyte complex formation using alginate and chitosan. Carbohydrate
poly-siRNA/glycol chitosan self-cross-linked nanoparticles for systemic siRNA de- Polymers, 74, 813–821.
livery in cancer treatment. Angewandte Chemie International Edition, 51, 7203–7207. Sarmento, & Bruno (2012). Chitosan-based systems for biopharmaceuticals. John Wiley &
Lee, S. J., Min, H. S., Ku, S. H., Son, S., Kwon, I. C., Kim, S. H., et al. (2014). Tumor- Sons.
targeting glycol chitosan nanoparticles as a platform delivery carrier in cancer di- Sato, T., Ishii, T., & Okahata, Y. (2001). In vitro gene delivery mediated by chitosan.
agnosis and therapy. Nanomedicine Nanotechnology Biology and Medicine, 9, Effect of pH, serum, and molecular mass of chitosan on the transfection efficiency.
1697–1713. Biomaterials, 22, 2075–2080.
Lee, S. J., Yook, S., Yhee, J. Y., Yoon, H. Y., Kim, M.-G., Ku, S. H., et al. (2015). Co- Shi, G.-N., Zhang, C.-N., Xu, R., Niu, J.-F., Song, H.-J., Zhang, X.-Y., et al. (2017).
delivery of VEGF and Bcl-2 dual-targeted siRNA polymer using a single nanoparticle Enhanced antitumor immunity by targeting dendritic cells with tumor cell lysate-
for synergistic anti-cancer effects in vivo. Journal of Controlled Release, 220, 631–641. loaded chitosan nanoparticles vaccine. Biomaterials, 113, 191–202.
Li, H., Huo, M., Zhou, J., Dai, Y., Deng, Y., Shi, X., et al. (2010). Enhanced oral absorption Siddiqui, I. A., Bharali, D. J., Nihal, M., Adhami, V. M., Khan, N., Chamcheu, J. C., et al.
of paclitaxel in N-deoxycholic acid-N, O-hydroxyethyl chitosan micellar system. (2014). Excellent anti-proliferative and pro-apoptotic effects of (−)-epigallocatechin-
Journal of Pharmaceutical Sciences, 99, 4543–4553. 3-gallate encapsulated in chitosan nanoparticles on human melanoma cell growth
Li, J., Huang, P., Chang, L., Long, X., Dong, A., Liu, J., et al. (2013). Tumor targeting and both in vitro and in vivo. Nanomedicine Nanotechnology Biology and Medicine, 10,
pH-responsive polyelectrolyte complex nanoparticles based on hyaluronic acid-pa- 1619–1626.
clitaxel conjugates and Chitosan for oral delivery of paclitaxel. Macromolecular Smith, A., Perelman, M., & Hinchcliffe, M. (2014). Chitosan: A promising safe and im-
Research, 21, 1331–1337. mune-enhancing adjuvant for intranasal vaccines. Human Vaccines &
Li, Y., Yang, J., Xu, B., Gao, F., Wang, W., & Liu, W. (2015). Enhanced therapeutic siRNA Immunotherapeutics, 10, 797–807.
to tumor cells by a pH-sensitive agmatine-chitosan bioconjugate. ACS Applied Son, Y. J., Jang, J.-S., Cho, Y. W., Chung, H., Park, R.-W., Kwon, I. C., et al. (2003).
Materials & Interfaces, 7, 8114–8124. Biodistribution and anti-tumor efficacy of doxorubicin loaded glycol-chitosan na-
Li, Y., Zheng, X., Zhang, X., Liu, S., Pei, Q., Zheng, M., et al. (2017). Porphyrin-based noaggregates by EPR effect. Journal of Controlled Release, 91, 135–145.
carbon dots for photodynamic therapy of hepatoma. Advanced Healthcare Materials, 6, Su, Y., Hu, Y., Du, Y., Huang, X., He, J., You, J., et al. (2015). Redox-responsive polymer-
1600924. drug conjugates based on doxorubicin and chitosan oligosaccharide-g-stearic acid for
Liao, Z.-X., Ho, Y.-C., Chen, H.-L., Peng, S.-F., Hsiao, C.-W., & Sung, H.-W. (2010). cancer therapy. Molecular Pharmaceutics, 12, 1193–1202.
Enhancement of efficiencies of the cellular uptake and gene silencing of chitosan/ Tan, Y.-l., & Liu, C.-G. (2009). Self-aggregated nanoparticles from linoleic acid modified
siRNA complexes via the inclusion of a negatively charged poly(γ-glutamic acid). carboxymethyl chitosan: Synthesis, characterization and application in vitro. Colloids
Biomaterials, 31, 8780–8788. and Surfaces B, Biointerfaces, 69, 178–182.
Liu, J., Liang, H., Li, M., Luo, Z., Zhang, J., Guo, X., et al. (2018). Tumor acidity activating Tapola, N. S., Lyyra, M. L., Kolehmainen, R. M., Sarkkinen, E. S., & Schauss, A. G. (2008).

11
E. Zhang, et al. Carbohydrate Polymers 222 (2019) 115004

Safety aspects and cholesterol-lowering efficacy of chitosan tablets. Journal of the Xu, Y., Wang, L., Li, Y.-K., & Wang, C.-Q. (2014). Oxidation and pH responsive nano-
American College of Nutrition, 27, 22–30. particles based on ferrocene-modified chitosan oligosaccharide for 5-fluorouracil
Tian, Q., Zhang, C.-N., Wang, X.-H., Wang, W., Huang, W., Cha, R.-T., et al. (2010). delivery. Carbohydrate Polymers, 114, 27–35.
Glycyrrhetinic acid-modified chitosan/poly(ethylene glycol) nanoparticles for liver- Yang, C., Gao, S., & Kjems, J. (2014). Folic acid conjugated chitosan for targeted delivery
targeted delivery. Biomaterials, 31, 4748–4756. of siRNA to activated macrophages in vitro and in vivo. Journal of Materials Chemistry
Tsai, W.-H., Yu, K.-H., Huang, Y.-C., & Lee, C.-I. (2018). EGFR-targeted photodynamic B, 2, 8608–8615.
therapy by curcumin-encapsulated chitosan/TPP nanoparticles. International Journal Yang, H., Chen, Y., Chen, Z., Geng, Y., Xie, X., Shen, X., et al. (2017). Chemo-photo-
of Nanomedicine, 13, 903–916. dynamic combined gene therapy and dual-modal cancer imaging achieved by pH-
United States Pharmacopoeia Convention (2016). The United States pharmacopoeia 40 responsive alginate/chitosan multilayer-modified magnetic mesoporous silica nano-
(USP40), the national formulary 35 (NF35). 7601–7605 Rockville: MD. composites. Biomaterials Science, 5, 1001–1013.
Wang, H., Di, J., Sun, Y., Fu, J., Wei, Z., Matsui, H., et al. (2015). Biocompatible PEG- Yang, W., Fu, J., Wang, T., & He, N. (2009). Chitosan/sodium tripolyphosphate nano-
chitosan@carbon dots hybrid nanogels for two-photon fluorescence imaging, near- particles: Preparation, characterization and application as drug carrier. Journal of
infrared light/pH dual-responsive drug carrier, and synergistic therapy. Advanced Biomedical Nanotechnology, 5, 591–595.
Functional Materials, 25, 5537–5547. Yin, H., Kanasty, R. L., Eltoukhy, A. A., Vegas, A. J., Dorkin, J. R., & Anderson, D. G.
Wang, J., Tao, X., Zhang, Y., Wei, D., & Ren, Y. (2010). Reversion of multidrug resistance (2014). Non-viral vectors for gene-based therapy. Nature Reviews Genetics, 15, 541.
by tumor targeted delivery of antisense oligodeoxynucleotides in hydroxypropyl- Yoon, H. Y., Son, S., Lee, S. J., You, D. G., Yhee, J. Y., Park, J. H., et al. (2014). Glycol
chitosan nanoparticles. Biomaterials, 31, 4426–4433. chitosan nanoparticles as specialized cancer therapeutic vehicles: Sequential delivery
Wang, X.-Y., Zhang, L., Wei, X.-H., & Wang, Q. (2013). Molecular dynamics of paclitaxel of doxorubicin and Bcl-2 siRNA. Scientific Reports, 4, 6878.
encapsulated by salicylic acid-grafted chitosan oligosaccharide aggregates. Yu, J., Xie, X., Wu, J., Liu, Y., Liu, P., Xu, X., et al. (2013). Folic acid conjugated glycol
Biomaterials, 34, 1843–1851. chitosan micelles for targeted delivery of doxorubicin: Preparation and preliminary
Wang, X., Chen, Y., Dahmani, F. Z., Yin, L., Zhou, J., & Yao, J. (2014). Amphiphilic evaluation in vitro. Journal of Biomaterials Science Polymer Edition, 24, 606–620.
carboxymethyl chitosan-quercetin conjugate with P-gp inhibitory properties for oral Zhang, B.-F., Xing, L., Cui, P.-F., Wang, F.-Z., Xie, R.-L., Zhang, J.-L., et al. (2015).
delivery of paclitaxel. Biomaterials, 35, 7654–7665. Mitochondria apoptosis pathway synergistically activated by hierarchical targeted
Wang, X., Zhen, X., Wang, J., Zhang, J., Wu, W., & Jiang, X. (2013). Doxorubicin delivery nanoparticles co-delivering siRNA and lonidamine. Biomaterials, 61, 178–189.
to 3D multicellular spheroids and tumors based on boronic acid-rich chitosan na- Zhang, C. G., Zhu, W. J., Liu, Y., Yuan, Z. Q., Yang, S. D., Chen, W. L., et al. (2016). Novel
noparticles. Biomaterials, 34, 4667–4679. polymer micelle mediated co-delivery of doxorubicin and P-glycoprotein siRNA for
Wang, Y., Yang, X., Yang, J., Wang, Y., Chen, R., Wu, J., et al. (2011). Self-assembled reversal of multidrug resistance and synergistic tumor therapy. Scientific Reports, 6,
nanoparticles of methotrexate conjugated O-carboxymethyl chitosan: Preparation, 23859.
characterization and drug release behavior in vitro. Carbohydrate Polymers, 86, Zhang, E., Xing, R., Liu, S., Li, K., Qin, Y., Yu, H., et al. (2019). Vascular targeted chitosan-
1665–1670. derived nanoparticles as docetaxel carriers for gastric cancer therapy. International
Weecharangsan, W., Opanasopit, P., Ngawhirunpat, T., Apirakaramwong, A., Rojanarata, Journal of Biological Macromolecules, 126, 662–672.
T., Ruktanonchai, U., et al. (2008). Evaluation of chitosan salts as non-viral gene Zhang, E. H., Xing, R. E., Liu, S., Li, K. C., Qin, Y. K., Yu, H. H., et al. (2017). Comparison
vectors in CHO-K1 cells. International Journal of Pharmaceutics, 348, 161–168. in docetaxel-loaded nanoparticles based on three different carboxymethyl chitosans.
Wei, W., Lv, P. P., Chen, X. M., Yue, Z. G., Fu, Q., Liu, S. Y., et al. (2013). Codelivery of International Journal of Biological Macromolecules, 101, 1012–1018.
mTERT siRNA and paclitaxel by chitosan-based nanoparticles promoted synergistic Zhang, Y., Huo, M., Zhou, J., Yu, D., & Wu, Y. (2009). Potential of amphiphilically
tumor suppression. Biomaterials, 34, 3912–3923. modified low molecular weight chitosan as a novel carrier for hydrophobic antic-
Wicki, A., Witzigmann, D., Balasubramanian, V., & Huwyler, J. (2014). Nanomedicine in ancer drug: Synthesis, characterization, micellization and cytotoxicity evaluation.
cancer therapy: Challenges, opportunities, and clinical applications. Journal of Carbohydrate Polymers, 77, 231–238.
Controlled Release, 200, 138–157. Zhang, Y., Lv, T., Zhang, H., Xie, X., Li, Z., Chen, H., et al. (2017). Folate and hepta-
Wilhelm, S., Tavares, A. J., & Chan, W. C. W. (2016). Reply to “Evaluation of nanome- methine cyanine modified chitosan-based nanotheranostics for tumor targeted near-
dicines: Stick to the basics”. Nature Reviews Materials, 1, 16074. infrared fluorescence imaging and photodynamic therapy. Biomacromolecules, 18,
Wilhelm, S., Tavares, A. J., Dai, Q., Ohta, S., Audet, J., Dvorak, H. F., et al. (2016). 2146–2160.
Analysis of nanoparticle delivery to tumours. Nature Reviews Materials, 1, 16014. Zhu, Y., Wen, L., Shao, S., Tan, Y., Meng, T., Yang, X., et al. (2018). Inhibition of tumor-
Xu, Q., Guo, L., Gu, X., Zhang, B., Hu, X., Zhang, J., et al. (2012). Prevention of colorectal promoting stroma to enforce subsequently targeting AT1R on tumor cells by patho-
cancer liver metastasis by exploiting liver immunity via chitosan-TPP/nanoparticles logical inspired micelles. Biomaterials, 161, 33–46.
formulated with IL-12. Biomaterials, 33, 3909–3918.

12

You might also like