A Review of Biodegradable Polymeric Systems For Oral Insulin Delivery

You might also like

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

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

net/publication/278041012

A review of biodegradable polymeric systems for oral insulin delivery

Article  in  Drug Delivery · June 2015


DOI: 10.3109/10717544.2015.1052863 · Source: PubMed

CITATIONS READS

64 609

6 authors, including:

Xiang Yuan Xiong


Jiangxi Science & Technology Normal University
30 PUBLICATIONS   826 CITATIONS   

SEE PROFILE

All content following this page was uploaded by Xiang Yuan Xiong on 30 November 2015.

The user has requested enhancement of the downloaded file.


This article was downloaded by: [Xiang Yuan Xiong]
On: 07 August 2015, At: 02:11
Publisher: Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: 5 Howick Place,
London, SW1P 1WG

Drug Delivery
Publication details, including instructions for authors and subscription information:
http://www.tandfonline.com/loi/idrd20

A review of biodegradable polymeric systems for oral


insulin delivery
a a b a a a
Yue Yuan Luo , Xiang Yuan Xiong , Yuan Tian , Zi Ling Li , Yan Chun Gong & Yu Ping Li
a
School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, China
and
b
China National Pharmaceutical Industry Co., Ltd., Beijing, China
Published online: 24 Jun 2015.

Click for updates

To cite this article: Yue Yuan Luo, Xiang Yuan Xiong, Yuan Tian, Zi Ling Li, Yan Chun Gong & Yu Ping Li (2015): A review of
biodegradable polymeric systems for oral insulin delivery, Drug Delivery

To link to this article: http://dx.doi.org/10.3109/10717544.2015.1052863

PLEASE SCROLL DOWN FOR ARTICLE

Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained
in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no
representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the
Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and
are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and
should be independently verified with primary sources of information. Taylor and Francis shall not be liable for
any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever
or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of
the Content.

This article may be used for research, teaching, and private study purposes. Any substantial or systematic
reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any
form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http://
www.tandfonline.com/page/terms-and-conditions
http://informahealthcare.com/drd
ISSN: 1071-7544 (print), 1521-0464 (electronic)

Drug Deliv, Early Online: 1–10


! 2015 Informa Healthcare USA, Inc. DOI: 10.3109/10717544.2015.1052863

REVIEW ARTICLE

A review of biodegradable polymeric systems for oral insulin delivery


Yue Yuan Luo1, Xiang Yuan Xiong1, Yuan Tian2, Zi Ling Li1, Yan Chun Gong1, and Yu Ping Li1
1
School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, China and 2China National Pharmaceutical Industry Co., Ltd.,
Beijing, China

Abstract Keywords
Currently, repeated routine subcutaneous injections of insulin are the standard treatment for Bioavailability, biodegradable, insulin,
insulin-dependent diabetic patients. However, patients’ poor compliance for injections often oral delivery, polymer
fails to achieve the stable concentration of blood glucose. As a protein drug, the oral
bioavailability of insulin is low due to many physiological reasons. Several carriers, such as History
Downloaded by [Xiang Yuan Xiong] at 02:11 07 August 2015

macromolecules and liposomes have been used to deliver drugs in vivo. In this review article,
the gastrointestinal barriers of oral insulin administration are described. Strategies for Received 7 April 2015
increasing the bioavailability of oral insulin, such absorption enhancers, enzyme inhibitors, Revised 15 May 2015
enteric coatings are also introduced. The potential absorption mechanisms of insulin-loaded Accepted 15 May 2015
nanoparticles across the intestinal epithelium, including intestinal lymphatic route, transcellular
route and paracellular route are discussed in this review. Natural polymers, such as chitosan and
its derivates, alginate derivatives, g-PGA-based materials and starch-based nanoparticles have
been exploited for oral insulin delivery; synthetic polymers, such as PLGA, PLA, PCL and PEA
have also been developed for oral administration of insulin. This review focuses on recent
advances in using biodegradable natural and synthetic polymers for oral insulin delivery along
with their future prospects.

Introduction enhancers (Degim et al., 2004; Zhang et al., 2011a,b) to


enhance the absorption of insulin. But most of the recent
To be effective, an orally administered protein drug must
literatures concentrate on the novel drug delivery systems
transit along the gastrointestinal (GI) tract, pass through the
made of biodegradable macromolecular materials, which
mucus layer, traverse across the intestinal epithelium, enter
could be used to carry insulin. In this review, the attention has
into the portal vein and finally reach the systemic circulation
been focused on the use of biodegradable natural and
(Chen et al., 2011).
synthetic polymers (Alonso, 2004).
However, oral administration of pure insulin showed less
effect due to the following factors. Firstly, insulin can be
easily degraded by the acidic conditions in stomach and the The absorption of oral insulin
proteases existing in the GI tract. Secondly, insulin is a kind
After the oral administration of insulin preparations, the drugs
of hydrophilic protein with a molecular weight of about 6000
should firstly transport along the GI tract. However, the pH
Da, and thus it is difficult for insulin to be encapsulated in
of stomach environment is 1.2–3.0 and the pH in intestine is
hydrophobic carriers and the permeation of insulin through
6.5–8.0. Such variations make it difficult for protein drugs,
the intestinal epithelium is limited. Thirdly, because of the
such as insulin to remain activity in GI tract (Wang & Zhang,
liver first-pass effect, the bioavailability of untreated insulin is
2012). Some pH-sensitive materials, including hydroxypropyl
extremely low (Sonaje et al., 2010a,b,c).
methylcellulose phthalate (HPMCP) (Maroni et al., 2009;
In order to solve these problems, a lot of work has been
Wang & Zhang, 2012; Wu et al., 2012a,b,c), polycarboxylates,
carried out to develop new oral insulin delivery systems. The
polyaminomethacrylates (EudragitÕ ) (Wu et al., 2012a,b,c;
loading efficiency of insulin on hydrophobic carriers could be
Marais et al., 2013; Viehof et al., 2013) were used as enteric
improved by the addition of distearyldimethylammonium
coatings. A previous study showed that HP55 (one kind of
bromide or soybean phospholipid (Cui et al., 2006; Li et al.,
HPMCP) modifications were capable of protecting insulin
2013). Other studies used enteric coatings (Zhao et al., 2013),
against acidic gastrointestinal conditions and then dissolving in
enzyme inhibitors (Werle et al., 2007) and permeation
the small intestinal environment. This modification reduced
the cumulative release rate of insulin in simulated gastric fluid
Address for correspondence: Dr Xiang Yuan Xiong, School of Life from 33.24 to 15.87%, while cumulative release rates of insulin
Science, Jiangxi Science & Technology Normal University, Nanchang
330013, China. Tel: 86-791-8539361. Fax: 86-791-3815794. Email: in simulated intestinal fluid increased from 15 to 58.06% after
xyxiong@gmail.com this modification (Zhao et al., 2013).
2 Y. Y. Luo et al. Drug Deliv, Early Online: 1–10

In addition to the acidic environment, insulin could also be thus bypassing the first-pass effects and improving the
hydrolyzed by the pepsins and other proteases in the GI tract. bioavailability (Yun et al., 2013). Peyer’s patches act as an
In a previous study, trypsin/chymotrypsin inhibitor aprotinin entrance for drug to the lymphatic vessels, via the M cell
was used to protect insulin from degradation. Eight hours transportation of peptides from the intestinal lumen to the
after oral administration of insulin-loaded chitosan–aprotinin underlying lymphoid cells. Lymphatic pathway has been
conjugate, the mean blood glucose level decreased to shown to function in the absorption of highly lipophilic
84 ± 6%, while the mean blood glucose level in the control compounds. According to a previous study, the liposomal
group (rats received no treatment) increased to 121 ± 8% of formulation could facilitate the uptake of cefotaxime (a
the initial blood glucose level (Werle et al., 2007). However, hydrophilic drug) through lymphatic absorption. The drug
using enzyme inhibitors may lead to serious side effects, such concentration in the lymph was higher than that in the plasma
as pancreatic hypertrophy, impairs dietary protein digestion when administered via liposomal formulation. The bioavail-
and some potentially harmful changes in body functions ability of oral cefotaxime-loaded liposomes increased about
(Rekha & Sharma, 2013). 2.7 and 2.3 times compared with its oral administration via
Before traveling across the intestinal epithelium, the drugs aqueous solution and the physical mixture, respectively (Ling
should firstly pass through the intestinal mucus layer. As a et al., 2006).
selective barrier which enables the exchange of nutrients,
water, and small molecules while being impermeable to Transcellular route
pathogens, mucus layer is negatively charged and renewed
continuously (Cone, 2009). Generally speaking, molecules Uptake of NPs via the transcellular pathway depends on their
with neutral surfaces could cross the mucus layer easily, while particle size and hydrophobicity. It is known that particles
Downloaded by [Xiang Yuan Xiong] at 02:11 07 August 2015

positively charged molecules have a stronger attraction with smaller than 300 nm could be uptaken by enterocytes (He et al.,
mucus (des Rieux et al., 2013). Some bioadhesive materials 2012), while particles larger than 500 nm were more likely to
were used to increase the intestinal retention time. Chitosan, a be absorbed in jejuna Peyer’s patches (Bakhru et al., 2013).
cationic polymer, was the most widely used due to its Han et al. (2012) used insulin-loaded hyaluronic acid (HA)
electrostatic interaction with negatively charged mucins (Sung nanoparticles for insulin delivery through transcellular route.
et al., 2012). Some weakly anionic carboxyl-containing The plasma glucose levels of diabetic rats receiving oral
polymers, including poly(acrylic acid), poly(methacrylic administration of insulin-loaded HA nanoparticles (50 IU/kg)
acid), carboxymethylcellulose, sodium alginate were capable reduced by 24% in 2 h and 32–39% in during 3–8 h, while the
of forming hydrogen-bonds with mucins by binding their group treated with the same dose of insulin solution showed no
carboxylic segments to the oligosaccharide branches of change in blood glucose (Han et al., 2012).
mucins, which contribute to their mucoadhesiveness In general, bioadhesive materials showed non-specific
(Khutoryanskiy, 2011). interactions with mucus layers. Receptor-mediated transport
via receptor-specific ligands modifications of insulin delivery
Three possible uptake routes in the small intestine carriers were used to enhance the absorption of oral insulin.
Promising targets, such as enterocytes, mucus layers and M
Mostly, there are three possible routes for the absorption of
cells have been studied (des Rieux et al., 2013). Biotin
nanoparticles-based oral insulin delivery system (Figure 1):
(vitamin B7) (Zhang et al., 2014) and lectins, such as wheat
the first, via the densely clustered M cells located on the germ agglutinin (WGA) (capable for binding to N-acetylglu-
surface of Peyer’s patches (Lopes et al., 2014); the second, via cosamine and sialic acid), tomato lectin, Ulex europaeus
transcellular route, which mainly absorbs lipophilic particles agglutinin1 have been exploited to modify the protein drug
through the cell membrane of enterocytes; and the last, via carriers (Kim et al., 2005; Zhang et al., 2005). Vitamin B12-
paracellular avenues, through the tight junctions (TJs) coated nanospheres could also be used as target delivery
between two adjacent enterocytes (Shahbazi & Santos, 2013). carrier through VB12-IF-IF receptor mediated endocytosis of
villous epithelium. Insulin-loaded dextran nanoparticles
Intestinal lymphatic route
conjugated with VB12 derivatives showed profound (70–
As an alternative way for oral insulin delivery, the intestinal 75% blood glucose reductions) and prolonged (54 h) anti-
lymphatic pathway transports drugs directly to the vena cava, diabetic effects in STZ-diabetic rats (Chalasani et al.,

Figure 1. Three possible routes through the


intestinal epithelium.
DOI: 10.3109/10717544.2015.1052863 Review of biodegradable polymeric systems for oral insulin delivery 3

2007a,b). Goblet cell-targeting ligands, such as CSKSSDYQC Other biocompatible polymers, including poly(acrylic
peptide, which identified from a random phage-peptide acid) and chitosan, are capable of opening the tight junctions
library was found to have affinity with the goblet cells. reversibly (Woitiski et al., 2011; Gao et al., 2013a,b).
After oral administration in diabetic rats, the CSKSSDYQC Poly(acrylic acid) and its derivatives have shown ability in
peptide modified insulin-loaded nanoparticles showed a better binding Ca2+ (Gao et al., 2013a,b). Oral administration of
hypoglycemic effect with a 1.5-fold higher relative bioavail- insulin-loaded carboxymethyl cellulose/poly(acrylic acid)
ability compared with unmodified ones (Jin et al., 2012). hydrogels (60 IU/kg) to diabetic rats reduced fasting plasma
glucose levels to 72.4% within 6 h post-administration. The
Paracellular route relative bioavailability of oral administration of the insulin-
As to hydrophilic molecules, the paracellular pathway is the loaded poly(acrylic acid) hydrogels (6.59%) was 10 times
only way for their uptake. However, the tight junctions are higher than oral administration of free insulin solution (less
difficult for large molecules to pass through. Drugs are than 0.5%) (Gao et al., 2013a,b). However, the opening of the
generally excluded from this pathway without the use of tight junctions might cause the absorption of bacterial toxins,
permeation enhancer (Chuang et al., 2013). As the decreasing leading to some safety problems (Chen et al., 2011). The
of calcium ion concentration leads to the opening of the tight approaches to improve the bioavailability of insulin are
junctions, Ca2+ chelating agents, such as ethylene glycol summarized in Table 1.
tetraacetic acid (EGTA) (Chuang et al., 2013), diethylene
triamine pentaacetic acid (DTPA) (Su et al., 2012), could Natural polymers
enhance the paracellular permeability by reversible opening
Chitosan
Downloaded by [Xiang Yuan Xiong] at 02:11 07 August 2015

of the tight junctions. Caco-2 cell monolayers treated with


EGTA or g PGA–EGTA nanoparticles produced a signifi- Chitosan (CS) is a natural polysaccharide derived from
cantly larger reduction in TEER compared to the control cells the partial deacetylation of chitin, a biopolymer generally
(Chuang et al., 2013). Since calcium ions are also very crucial found in crustacean shells and insects. The primary building
to sustain the activity of many intestinal proteases, Ca2+ blocks of CS are glucosamine and N-acetyl-glucosamine. It is
chelating agents could also be used as an enzyme inhibitor to biocompatible, biodegradable and protective (Mukhopadhyay
protect insulin against intestinal proteases. et al., 2013). The utilization of chitosan for oral insulin
Bile salts, such as sodium glycocholate (Niu et al., 2011), delivery is based on the mucoadhesive property and the
sodium taurocholate (Degim et al., 2004) and sodium ability to mediate the opening of the tight junctions reversibly
deoxycholate (Niu et al., 2012) were most widely used to (Jose et al., 2012, 2013) (Figure 2).
modify insulin-loaded liposomes as a penetration enhancer. A recently published report studied an iron oxide–CS
These might be attributed to the good deformability of nanocomposite loaded with insulin. The iron oxide nanopar-
liposomes containing bile salts (Hu et al., 2013). Niu et al. ticles were obtained in chitosan solution by eco-friendly laser
(2012) compared the differences between liposomes contain- ablation technique. The insulin-loaded iron oxide-chitosan
ing sodium glycocholate, sodium taurocholate and sodium nanoparticles were found to effectively reduce the blood
deoxycholate, and the highest oral bioavailability was observed glucose level when administered orally in diabetic rats. About
in rats administrated with insulin-loaded sodium glycocholate 51% blood glucose level reductions were achieved in severely
(8.5 and 11.0% in non-diabetic and diabetic rats, respectively). diabetic rats (Kebede et al., 2013). This approach is

Table 1. Approaches to improve the bioavailability of insulin.

Approaches Systems Mechanisms Shortcomings References


Insulin modifications Distearyldimethylammoni- Improve the hydrophobicity (Cui et al., 2006; Li et al.,
um bromide, soybean of insulin 2013)
phospholipid
Permeation enhancers Chelators (EGTA, DTPA) Chelating Ca2+, thus Cause the absorption of (Su et al., 2012; Chuang
opening the tight bacterial toxins in GI et al., 2013)
junctions tract
Bile salts Open the tight junctions (Degim et al., 2004;
and improve deformabil- Niu et al., 2011, 2012;
ity and permeation Hu et al., 2013)
Enteric coatings HPMCP, EudragitÕ pH-sensitive Rapid release in intestinal (Cui et al., 2007; Maroni
et al., 2009; Wang &
Zhang, 2012; Wu et al.
2012a,b,c; Marais et al.,
2013; Viehof et al.,
2013; Zhao et al., 2013)
Enzyme inhibitors Trypsin/chymotrypsin Protect insulin from deg- Introducing side effects or (Werle et al., 2007)
inhibitor radation by proteases in do harm to body
GI tract functions
Bioadhesive materials Chitosan, PAA, PMAA Adhere to mucus layer and Limited by the turnover of (Khutoryanskiy, 2011;
prolong the retention the mucus layer Sung et al., 2012)
time
4 Y. Y. Luo et al. Drug Deliv, Early Online: 1–10

Figure 2. Schematic illustrations of the opening of chitosan-mediated reversible tight junction (TJ) (Sung et al., 2012).
Downloaded by [Xiang Yuan Xiong] at 02:11 07 August 2015

promising, since the nanoparticles can be used in site-directed (Yin et al., 2009). When comparing the mucoadhesion of
drug delivery due to its magnetic properties. different thiolated chitosans, it was found that Chitosan–
thiobutylamidine exhibited the highest mucoadhesion, fol-
CS derivatives lowed by Chitosan–4-mercaptobenzoic acid, Chitosan–gluta-
The pKa of CS is approximately 6.5. CS is insoluble in thione, Chitosan–6-mercaptonicotinic acid, Chitosan–N-
neutral environment, where CS starts to lose charge, and this acetyl cysteine, Chitosan–thioglycolic acid and Unmodified
leads to the loss of its mucoadhesive properties and TJ chitosan (Mueller et al., 2011).
opening activities (Qian et al., 2006). To overcome these Previous research studied carboxylated chitosan by mod-
drawbacks, several CS derivatives, including quaternized ifying chitosan with negatively charged carboxyl groups, in
chitosan, thiolated chitosan, carboxylated chitosan and order to increase the water solubility of chitosan (Cui et al.,
amphiphilic chitosan have been synthesized and evaluated 2007; Liu et al., 2013). In a previous study, carboxylated
for their potential in oral insulin delivery (Chen et al., 2013). chitosan-grafted poly(methyl methacrylate) nanoparticles
Quaternized chitosan can reserve its positive charge in (CCGN) were prepared. The pharmacological bioavailability
neutral condition, which leads to a residence time increase after oral administration of insulin-loaded CCGN at 25 IU/kg
and enhanced bioavailability (Sun & Wan, 2007). Sonia et al. was 9.7%, while a long-lasting hypoglycemic effect could be
(2011) explored N-(2-hydroxyl) propyl-3-trimethyl ammo- observed after oral administration of insulin-loaded CCGN
nium chitosan chloride (HTCC) for oral insulin delivery. The (Cui et al., 2009).
results showed that quaternized chitosan had a stronger Amphiphilic CS derivatives, such as lauroyl sulphated
electrostatic interaction with mucus, and the percentage of chitosan (LSCS) were used for oral insulin delivery. It was
adhesion was 95% for HTCC (the percentage of free amino showed that LSCS were non-toxic and lauroyl sulfated
groups was 36%) as compared to 72% for naive chitosan (the modification on chitosan improves mucoadhesion of CS
percentage of free amino groups was 78%). However, the high greatly. Moreover, LSCS could also protect insulin from
positive charge of quaternized CS derivatives may cause enzymatic degradation and reversely open the tight junctions
toxicity to cell membranes. Poly(ethylene glycol) (PEG) was (Shelma & Sharma, 2011).
used to modify N-Trimethyl chitosan chloride (TMC) in order
Alginate derivatives
to reduce the cytotoxicity. The hemolysis assay showed that
TMC modified with PEG has a better biocompatibility than Alginate, an anionic mucoadhesive polysaccharide consist-
TMC without PEG modification (Prego et al., 2006; ing of various ratios of b-D-mannuronopyranosyl and
Zhu et al., 2007). a-L-guluronopyranosyl units linked by (1!4)-O-glycosidic
Thiolated chitosan have been proved to be more bonds, has always been used for preparing microparticles
mucoadhesive compared with unmodified CS (Chen et al., (Chan & Neufeld, 2010). It shows a property of mild gel-
2013). The thiolated TMC, trimethyl chitosan-cysteine formation by ionically cross-linked with multivalent cations,
(TMC-Cys) was prepared to combine the mucoadhesion of such as calcium ions, thus enabling drug retention within the
TMC and the permeation enhancing abilities of thiolated gel matrix (Sarmento et al., 2007a,b). The large porosity of
polymers for oral insulin delivery. Compared to TMC alginate beads often leads to low encapsulation efficiency and
nanoparticles, TMC-Cys nanoparticles significantly increased rapid release of the drug. Low drug encapsulation efficiency
insulin absorption through rat intestine by 1.7 to 2.6-fold, of alginates can be improved by the addition of chitosan and
and mucoadhesion and permeation enhancing effects of dextran sulfate (Martins et al., 2007; Reis et al., 2007; Sonia
TMC-Cys nanoparticles were improved significantly & Sharma, 2012).
DOI: 10.3109/10717544.2015.1052863 Review of biodegradable polymeric systems for oral insulin delivery 5

Alginate/chitosan capsules were widely used for controlled


release of oral protein drugs (Sarmento et al., 2007a,b; Čalija
et al., 2011; Zhang et al., 2011a,b). Zhang et al. (2011a,b)
encapsulated insulin in alginate–chitosan microspheres via
different loading methods, and it was shown that the highest
loading efficiency (56.7%) and remarkable activity mainten-
ance (99.4%) were achieved when the insulin was loaded
during the chitosan solidification process, while the network
formation between chitosan and alginate-Ca microspheres
greatly reduced the porosity and decreased the leakage of
insulin.

g-PGA-based materials
Poly-g-glutamic acid (g-PGA) is a water-soluble, biodegrad-
able and non-toxic polymer. Many studies used nanoparticles
composed of poly-g-glutamic acid (g-PGA) and CS for oral
insulin delivery. With a smaller size and a higher loading
efficiency compared with purely CS nanoparticles (Lin et al.,
2007), the pH-sensitive CS/g-PGA nanoparticles could resist
Downloaded by [Xiang Yuan Xiong] at 02:11 07 August 2015

gastric acid while released rapidly in certain area of the small


intestine (Sonaje et al., 2010a,b,c). g-PGA conjugated cova-
lently by diethylene triamine pentaacetic acid (DTPA) could
further prevent enzymolysis and prolong the residence time
of the CS/g-PGA-DTPA system for oral insulin delivery
(Su et al., 2012).
Tripolyphosphate (TPP) and MgSO4 were added into
CS/g-PGA nanoparticles to form an ionically cross-linked
network. Comparing with CS/g-PGA nanoparticles, the multi-
ion-cross-linked nanoparticles could retain more insulin. With
the addition of MgSO4 and TPP, the cumulative amount of Figure 3. (a) Blood glucose level versus time profiles and (b) plasma
insulin released were significantly reduced at pH 2.5 and pH insulin level versus time profiles of test diabetic rats following the
7.0, while a fast release of insulin was observed at pH 7.4 administration of different insulin formulations (Sonaje et al., 2010a,b,c).
(simulating the pH in the body fluid at intercellular spaces),
which means insulin were more likely to be released from the
multi-ion-cross-linked nanoparticles after they were infil- PGA chain. Insulin released from the copolymer in artificial
trated into mucus layer (Lin et al., 2008). Since SC injection gastric juice (pH ¼ 1.2) more slowly than that in artificial
of insulin often produces peripheral hyperinsulinemia in intestinal liquid (pH ¼ 6.8). In another study, hydrophobic
individuals. In another study, it was found that oral admin- starch acetate was conjugated with polyethylene glycol (PEG)
istration of aspart-insulin-loaded chitosan/g-PGA nanoparti- to from an amphiphilic polymeric derivative. Being pH-
cles could achieve a slower but longer hypoglycemic effect sensitive, the PEGylated starch acetate nanoparticles are
without producing hyperinsulinemia. Oral administration of capable of opening the tight junctions. Moreover, work of
aspart-insulin-loaded chitosan/g-PGA nanoparticles at insulin adhesion for PEGylated starch acetate nanoparticles was also
doses of 30 IU/kg produced a hypoglycemic effect similar to significantly higher at 595.7 mN mm than starch acetate
subcutaneous injections of neutral protamine Hagedorn (179.1 mN mm) and chitosan (144.4 mN mm), which means
insulin (NPH-insulin) at insulin doses of 5 IU/kg. Moreover, PEGylated starch acetate nanoparticles showed a high
the SC injection of aspart-insulin solution (5 IU/kg) produced mucoadhesiveness (Minimol et al., 2013).
a better hypoglycemic effect than SC injection of NPH-
insulin solution with the same dose (Sonaje et al., 2010a,b,c) Synthetic polymers
(Figure 3). Since the natural polymers come from different preparation
technologies and different sources, which may lead to a
Starch-based nanoparticles variety of physicochemical properties (Sonia & Sharma,
As one of the better known natural and biodegradable 2012). However, the structure and physicochemical properties
biopolymers, starch has attracted much attention because of of synthetic polymeric carriers could be well-controlled, and
its gelling and film forming properties. Zhang et al. (2013) thus their drug release properties and biological behaviors
obtained a pH-responsive copolymer made of starch nano- could be modified to change.
particles as backbone and poly(L-glutamic acid) (PGA) as
PLGA
graft chains. The result of in vitro insulin release experiment
showed that the grafted copolymer had excellent pH- Poly(lactide-co-glycolide) (PLGA) is widely studied for
responsive property due to the introduction of pH-responsive biological applications owing to their biodegradability and
6 Y. Y. Luo et al. Drug Deliv, Early Online: 1–10

Concanavalin A was used as a target ligand to enhance the


lymphatic absorption of oral insulin formulations. After oral
administration of Concanavalin A-conjugated PLGA nano-
particles (20 IU/kg), the blood glucose lever reduced from
about 260.17 to 196.33 mg/dL in 3 h, and hypoglycemic effect
lasted for the next 24 h, while oral PLGA nanoparticles loaded
with the same dose of insulin cannot reach the same effect
(Hurkat et al., 2012).

PLA
Vesicles self-assembled from amphiphilic polymers are cap-
able of encapsulating both hydrophilic and hydrophobic drugs
due to their hydrophilic shell and hydrophobic core in aqueous
solutions (Kita-Tokarczyk et al., 2005; Xiong, 2006). Xiong
et al. explored the use of Pluronic/poly(lactic acid) (PLA)
vesicles as oral insulin carriers. Pluronic/PLA copolymer was
obtained by attaching PLA segments to both ends of Pluronic
copolymer by a ring-opening polymerization (Figure 5). PLA
was highly hydrophobic, while the hydrophobicity of PPO was
Downloaded by [Xiang Yuan Xiong] at 02:11 07 August 2015

weaker and PEO was hydrophilic, so the Pluronic/PLA


copolymer could self-assemble to vesicles in water (Figure 6).
According to their studies, after orally treating with
insulin-loaded PLA-F127-PLA vesicles at insulin doses of
50 IU/kg, the blood glucose of the diabetic mice reduced from
18.5 to 5.3 mmol within 4.5 h, and this effect lasted for an
additional 18.5 h (Xiong et al., 2007). Pluronic P85/PLA
(PLA-P85-PLA) vesicles were also explored to carry insulin
for oral administration. After oral administration of insulin-
loaded PLA-P85-PLA vesicles at insulin doses of 200 IU/kg
to the diabetic mice, the glucose level reached the minimum
Figure 4. (a) Short model of PLGA. (b) Interaction of the insulin with a after 2.5 h. The blood glucose level increased slowly after
PLGA model of near 1500 Da. In this figure the histidine are shown in 10.5 h and was maintained at a low level (32.5% of initial
space filling, the glutamate in ball and stick (Lassalle & Ferreira, 2010).
blood glucose concentration) for at least an additional 14 h
(Figure 7) (Xiong et al., 2013).
biocompatibility (Cui et al., 2007; Lassalle & Ferreira 2010;
Reix et al., 2012). Generally speaking, the PLGA copolymers
were synthesized from lactic acid and glycolic acid through PCL
the ring-opening polymerization. The hydrophobic inter- Poly("-caprolactone) is a biodegradable polyester synthesized
actions between insulin and PLGA appear to be responsible from"-caprolactone by a ring-opening polymerization
for the entrapment of insulin in PLGA nanoparticles (Lassalle (Jugminder & Chawla, 2002). Polymeric nanoparticles
& Ferreira, 2010). The contact between insulin and the short composed of biodegradable poly("-caprolactone) and poly-
model of PLGA is illustrated in Figure 4. However, the cationic non-biodegradable acrylic polymer (EudragitÕ RS)
hydrophilicity of insulin makes them incapable of being were used for oral regular human insulin and aspart-insulin
entrapped by hydrophobic PLGA matrices. In a previous delivery. When aspart-insulin-loaded PCL/EudragitÕ RS
study, insulin was encapsulated in PLGA nanoparticles via nanoparticles were administered orally to diabetic rats at the
double-emulsion solvent evaporation method to improve the dose of 50 IU/kg, the maximum plasma glucose level
loading capacity. 11.31% insulin loading capacity was reduction (52%) was observed at 8 h after administration
obtained for the insulin-loaded PLGA nanoparticles. After (Damge et al., 2010). Comparing with oral administration of
oral administration of nanoparticles with an insulin concen- regular insulin-loaded PCL/EudragitÕ RS nanoparticles,
tration of 30 mg/kg to diabetic rats, a 50% reduction in blood which working only for 6–8 h, the hypoglycemic effects of
glucose level was observed within 8 h (Yang et al., 2012). aspart-insulin-loaded PCL nanoparticles lasted for 12–24 h
However, the slight negative surface charge of PLGA (Damge et al., 2007). This may be because the monomeric
nanoparticles exhibit weaker bioadhesive abilities compared aspart-insulin can be taken up by the intestinal mucosa more
with positive-charged nanoparticles. Cationic modifications, easily than regular human insulin (Damge et al., 2010).
such as chitosan-coat had been used to improve the bioavail-
ability of insulin-loaded PLGA nanoparticles. As a result,
PEA
compared with the PLGA nanoparticles, CS-coated PLGA
nanoparticles reduced the initial burst, and showed a stronger L -Lysine/L -Leucine-based poly(ester amide) with pendant
mucosal adhesion, thus prolonged the resistance time and COOH groups was synthesized by He et al. (2012) for oral
increased the bioavailability of insulin (Zhang et al., 2012). insulin delivery. The PEA copolymer with benzyl ester
DOI: 10.3109/10717544.2015.1052863 Review of biodegradable polymeric systems for oral insulin delivery 7
Figure 5 The synthesis scheme of
PLA-P85-PLA block copolymer (Xiong
et al., 2013).
Downloaded by [Xiang Yuan Xiong] at 02:11 07 August 2015

Figure 6. (a) TEM micrograph of PLA-P85-PLA nanoparticle vesicles in water. (b) The possible schematic microstructure of PLA-P85-PLA vesicles
(Xiong et al., 2013).

pendants was prepared by solution polycondensation of three


monomers, and the structure of L-Lysine/L-Leucine-based
PEA-COOH is illustrated in Figure 8 (He et al., 2012). Insulin
was encapsulated in PEA microspheres by a solid-in-oil-in-oil
technique using N,N-dimethyformamide(DMF)/corn oil as
solvent pairs. As a result, the release of insulin was pH-
dependent and the hydrophobicity of PEA-COOH micro-
spheres can be adjusted by the level of leucine components,
thus improving the stability of the drug delivery system and
enhancing the overall absorption of insulin. Oral administra-
tion of insulin-loaded PEA-COOH microspheres to strepto-
zotocin-induced diabetic rats at a dose of 60 IU/kg, the plasma
glucose levels reduced to 49.4% within the first 5 h of
administration, and this effect lasted for 8 h (He et al., 2012).
Arginine-based PEA (Arg-PEA) was also introduced in their
research; the structure of Arg-PEA is shown in Figure 8.
By adding Arg-PEA to form PEA-COOH/Arg-PEA blend Figure 7. The blood glucose level versus time profiles following oral
administration of physiological saline to control animals, insulin free
microspheres, the hypoglycemic effects could be further PLA-P85-PLA vesicles, insulin-loaded PLA-P85-PLA vesicles (50, 100
improved (He et al., 2013). and 200 IU/kg), and subcutaneous injection of free insulin (5 IU/kg).
Results are means ± S.D. of five animals per group. Significant
difference from negative physiological saline control (*p50.05;
Conclusions and future perspectives **p50.01) (Xiong et al., 2013).

Though oral insulin delivery systems show promise in recent


years, this work needs further study before clinical testing. improved via different method (Li et al., 2013). However, the
Since insulin is a hydrophilic carrier, the insulin has been bioavailability of orally administered insulin remains low
modified to enhance its physicochemical and biological compared with SC injections. In addition, maintaining the
stability. The loading efficiency of insulin should also be physicochemical and biological stability of insulin in the GI
8 Y. Y. Luo et al. Drug Deliv, Early Online: 1–10

Figure 8 Chemical structures of the poly(ester


amide)s. (a) L-Lysine/L-Leucine-based
PEA-COOH. (b) Arginine-based Arg-PEA
(He et al., 2013).

tract is necessary, especially when mucoadhesive systems Chuang EY, Lin KJ, Su FY, et al. (2013). Calcium depletion-mediated
protease inhibition and apical-junctional-complex disassembly via an
were used to prolong the residence time (Wong, 2010). EGTA-conjugated carrier for oral insulin delivery. J Control Release
Furthermore, potential insulin alternatives, such as psyllium 169:296–305.
polysaccharides (Singh & Chauhan, 2009), chalcone (Hsieh Cone RA. (2009). Barrier properties of mucus. Adv Drug Delivery Rev
et al., 2012), glucagon-like peptide-1 (Ahn et al., 2013), can 61:75–85.
Downloaded by [Xiang Yuan Xiong] at 02:11 07 August 2015

Cui F, He C, Yin L, et al. (2007). Nanoparticles incorporated in


be added to the oral dosage form in order to avoid the risk of bilaminated films: a smart drug delivery system for oral formulations.
insulin overdose. A successful oral insulin delivery system Biomacromolecules 8:2845–50.
should be biocompatible and biodegradable, keeping the Cui F, Shi K, Zhang L, et al. (2006). Biodegradable nanoparticles loaded
stability of insulin during the absorption, and ultimately with insulin-phospholipid complex for oral delivery: preparation,
in vitro characterization and in vivo evaluation. J Control Release 114:
improving the bioavailability of insulin. 242–50.
Cui FD, Tao AJ, Cun DM, et al. (2007). Preparation of insulin
loaded PLGA-Hp55 nanoparticles for oral delivery. J Pharm Sci 96:
Declaration of interest 421–7.
Cui FY, Qian F, Zhao Z, et al. (2009). Preparation, characterization, and
The authors would like to acknowledge the financial support oral delivery of insulin loaded carboxylated chitosan grafted
from the National Natural Science Foundation of China poly(methyl methacrylate) nanoparticles. Biomacromolecules 10:
(No. 21264009 and 31360376), the Natural Science 1253–8.
Foundation of Jiangxi Province (No. 20132BAB206034), the Damge C, Maincent P, Ubrich N. (2007). Oral delivery of insulin
associated to polymeric nanoparticles in diabetic rats. J Control
Scientific and Technological Landing Project of Higher
Release 117:163–70.
Education of Jiangxi Province (No. KJLD13071) and the Damge C, Socha M, Ubrich N, et al. (2010). Poly(epsilon-caprolactone)/
Cultivation Project for Academic and Technical Leaders of eudragit nanoparticles for oral delivery of aspart-insulin in the
Major Disciplines of Jiangxi Province.(No. 20153BCB22009). treatment of diabetes. J Pharm Sci 99:879–89.
Degim Z, Unal N, Essiz D, et al. (2004). The effect of various liposome
formulations on insulin penetration across Caco-2 cell monolayer. Life
References Sci 75:2819–27.
des Rieux A, Pourcelle V, Cani PD, et al. (2013). Targeted nanoparticles
Ahn S, Lee IH, Lee E, et al. (2013). Oral delivery of an anti-diabetic with novel non-peptidic ligands for oral delivery. Adv Drug Deliv Rev
peptide drug via conjugation and complexation with low molecular 65:833–44.
weight chitosan. J Control Release 170:226–32. Gao X, Cao Y, Song X, et al. (2013a). Biodegradable, pH-responsive
Alonso MJ. (2004). Nanomedicines for overcoming biological barriers. carboxymethyl cellulose/poly(acrylic acid) hydrogels for oral insulin
Biomed Pharmacother 58:168–72. delivery. Macromol Biosci 14:1–11.
Bakhru SH, Furtado S, Morello AP, et al. (2013). Oral delivery of Gao X, He C, Xiao C, et al. (2013b). Biodegradable pH-
proteins by biodegradable nanoparticles. Adv Drug Delivery Rev 65: responsive polyacrylic acid derivative hydrogels with tunable
811–21. swelling behavior for oral delivery of insulin. Polymer 54:
Čalija B, Cekić N, Savić S, et al. (2011). An investigation of formulation 1786–93.
factors affecting feasibility of alginate-chitosan microparticles for oral Han L, Zhao Y, Yin L, et al. (2012). Insulin-loaded pH-sensitive
delivery of naproxen. Arch Pharmacal Res 34:919–29. hyaluronic acid nanoparticles enhance transcellular delivery. AAPS
Chalasani KB, Russell-Jones GJ, Jain AK, et al. (2007a). Effective oral PharmSciTech 13:836–45.
delivery of insulin in animal models using vitamin B12-coated dextran He C, Yin L, Tang C, et al. (2012). Size-dependent absorption
nanoparticles. J Control Release 122:141–50. mechanism of polymeric nanoparticles for oral delivery of protein
Chalasani KB, Russell-Jones GJ, Yandrapu SK, et al. (2007b). A novel drugs. Biomaterials 33:8569–78.
vitamin B12-nanosphere conjugate carrier system for peroral delivery He P, Liu H, Tang Z, et al. (2013). Poly(ester amide) blend microspheres
of insulin. J Control Release 117:421–9. for oral insulin delivery. Int J Pharm 455:259–66.
Chan AW, Neufeld RJ. (2010). Tuneable semi-synthetic network alginate He P, Tang Z, Lin L, et al. (2012). Novel biodegradable and pH-sensitive
for absorptive encapsulation and controlled release of protein poly(ester amide) microspheres for oral insulin delivery. Macromol
therapeutics. Biomaterials 31:9040–7. Biosci 12:547–56.
Chen MC, Mi FL, Liao ZX, et al. (2013). Recent advances in chitosan- Hsieh CT, Hsieh TJ, El-Shazly M, et al. (2012). Synthesis of chalcone
based nanoparticles for oral delivery of macromolecules. Adv Drug derivatives as potential anti-diabetic agents. Bioorg Med Chem Lett
Deliv Rev 65:865–79. 22:3912–15.
Chen MC, Sonaje K, Chen KJ, et al. (2011). A review of the prospects Hu S, Niu M, Hu F, et al. (2013). Integrity and stability of oral liposomes
for polymeric nanoparticle platforms in oral insulin delivery. containing bile salts studied in simulated and ex vivo gastrointestinal
Biomaterials 32:9826–38. media. Int J Pharm 441:693–700.
DOI: 10.3109/10717544.2015.1052863 Review of biodegradable polymeric systems for oral insulin delivery 9
Hurkat P, Jain A, Jain A, et al. (2012). Concanavalin A Prego C, Torres D, Fernandez-Megia E, et al. (2006). Chitosan-PEG
conjugated biodegradable nanoparticles for oral insulin delivery. nanocapsules as new carriers for oral peptide delivery. Effect of
J Nanopart Res. doi: http://dx.doi.org/10.1007/s11051-012-1219-4. chitosan pegylation degree. J Control Release 111:299–308.
Jin Y, Song Y, Zhu X, et al. (2012). Goblet cell-targeting nanoparticles Qian F, Cui FY, Ding J, et al. (2006). Chitosan graft copolymer
for oral insulin delivery and the influence of mucus on insulin nanoparticles for oral protein drug delivery: preparation and charac-
transport. Biomaterials 33:1573–82. terization. Biomacromolecules 7:2722–7.
Jose S, Fangueiro JF, Smitha J, et al. (2012). Cross-linked chitosan Reis CP, Ribeiro AJ, Houng S, et al. (2007). Nanoparticulate delivery
microspheres for oral delivery of insulin: taguchi design and in vivo system for insulin: design, characterization and in vitro/in vivo
testing. Colloids Surf B 92:175–9. bioactivity. Eur J Pharm Biopharm 30:392–7.
Jose S, Fangueiro JF, Smitha J, et al. (2013). Predictive modeling of Reix N, Parat A, Seyfritz E, et al. (2012). In vitro uptake evaluation in
insulin release profile from cross-linked chitosan microspheres. Eur J Caco-2 cells and in vivo results in diabetic rats of insulin-loaded
Med Chem 60:249–53. PLGA nanoparticles. Int J Pharm 437:213–20.
Jugminder S, Chawla MMA. (2002). Biodegradable poly(o-caprolac- Rekha MR, Sharma CP. (2013). Oral delivery of therapeutic protein/
tone) nanoparticles for tumor-targeted delivery of tamoxifen. Int J peptide for diabetes – future perspectives. Int J Pharm 440:48–62.
Pharm 249:127–38. Sarmento B, Ribeiro A, Veiga F, et al. (2007a). Alginate/chitosan
Kebede A, Singh AK, Rai PK, et al. (2013). Controlled synthesis, nanoparticles are effective for oral insulin delivery. Pharm Res 24:
characterization, and application of iron oxide nanoparticles for oral 2198–206.
delivery of insulin. Lasers Med Sci 28:579–87. Sarmento B, Ribeiro AJ, Veiga F, et al. (2007b). Insulin-loaded
Khutoryanskiy VV. (2011). Advances in mucoadhesion and mucoadhe- nanoparticles are prepared by alginate ionotropic pre-gelation fol-
sive polymers. Macromol Biosci 11:748–64. lowed by chitosan polyelectrolyte complexation. J Nanosci
Kim BY, Jeong JH, Park K, et al. (2005). Bioadhesive interaction and Nanotechnol 7:2833–41.
hypoglycemic effect of insulin-loaded lectin-microparticle conjugates Shahbazi MA, Santos HA. (2013). Improving oral absorption via drug-
in oral insulin delivery system. J Control Release 102:525–38. loaded nanocarriers: absorption mechanisms, intestinal models and
Kita-Tokarczyk K, Grumelard J, Haefele T, et al. (2005). Block rational fabrication. Currt Drug Metab 14:28–56.
copolymer vesicles – using concepts from polymer chemistry to Shelma R, Sharma CP. (2011). Submicroparticles composed of amphi-
Downloaded by [Xiang Yuan Xiong] at 02:11 07 August 2015

mimic biomembranes. Polymer 46:3540–63. philic chitosan derivative for oral insulin and curcumin release
Lassalle V, Ferreira ML. (2010). PLGA based drug delivery systems applications. Colloids Surf B 88:722–8.
(DDS) for the sustained release of insulin: insight into the protein/ Singh B, Chauhan N. (2009). Modification of psyllium polysaccharides
polyester interactions and the insulin release behavior. J Chem for use in oral insulin delivery. Food Hydrocolloid 23:928–35.
Technol Biotechnol 85:1588–96. Sonaje K, Chen YJ, Chen HL, et al. (2010a). Enteric-coated capsules
Li P, Nielsen HM, Fano M, et al. (2013). Preparation and characteriza- filled with freeze-dried chitosan/poly(gamma-glutamic acid) nano-
tion of insulin-surfactant complexes for loading into lipid-based drug particles for oral insulin delivery. Biomaterials 31:3384–94.
delivery systems. J Pharm Sci 102:2689–98. Sonaje K, Lin KJ, Wang JJ, et al. (2010b). Self-assembled pH-sensitive
Lin YH, Chen CT, Liang HF, et al. (2007). Novel nanoparticles for oral nanoparticles: a platform for oral delivery of protein drugs. Adv Funct
insulin delivery via the paracellular pathway. Nanotechnology 18: Mater 20:3695–700.
1–11. Sonaje K, Lin KJ, Wey SP, et al. (2010c). Biodistribution, pharmaco-
Lin YH, Sonaje K, Lin KM, et al. (2008). Multi-ion-crosslinked dynamics and pharmacokinetics of insulin analogues in a rat model:
nanoparticles with pH-responsive characteristics for oral delivery of oral delivery using pH-responsive nanoparticles vs. subcutaneous
protein drugs. J Control Release 132:141–19. injection. Biomaterials 31:6849–58.
Ling SS, Magosso E, Khan NA, et al. (2006). Enhanced oral Sonia TA, Sharma CP. (2011). In vitro evaluation of N-(2-hydroxy)
bioavailability and intestinal lymphatic transport of a hydrophilic propyl-3-trimethyl ammonium chitosan for oral insulin delivery.
drug using liposomes. Drug Dev Ind Pharm 32:335–45. Carbohydr Polym 84:103–9.
Liu Y, Kong M, Feng C, et al. (2013). Biocompatibility, cellular uptake Sonia TA, Sharma CP. (2012). An overview of natural polymers for oral
and biodistribution of the polymeric amphiphilic nanoparticles as oral insulin delivery. Drug Discov Today 17:784–92.
drug carriers. Colloids Surf B 103:345–53. Su FY, Lin KJ, Sonaje K, et al. (2012). Protease inhibition and
Lopes MA, Abrahim BA, Cabral LM, et al. (2014). Intestinal absorption absorption enhancement by functional nanoparticles for effective oral
of insulin nanoparticles: contribution of M cells. Nanomedicine 10: insulin delivery. Biomaterials 33:2801–11.
1139–51. Sun Y, Wan A. (2007). Preparation of nanoparticles composed of
Marais E, Hamman J, Plessis L, et al. (2013). Eudragit(R) L100/N- chitosan and its derivatives as delivery systems for macromolecules.
trimethylchitosan chloride microspheres for oral insulin delivery. J Appl Polym Sci 105:552–61.
Molecules 18:6734–47. Sung HW, Sonaje K, Liao ZX, et al. (2012). pH-Responsive
Maroni A, Del Curto MD, Serratoni M, et al. (2009). Feasibility, stability nanoparticles shelled with chitosan for oral delivery of insulin:
and release performance of a time-dependent insulin delivery system from mechanism to therapeutic applications. Acc Chem Res 45:
intended for oral colon release. Eur J Pharm Biopharm 72:246–51. 619–29.
Martins S, Sarmento B, Souto EB, et al. (2007). Insulin-loaded alginate Viehof A, Javot L, Beduneau A, et al. (2013). Oral insulin delivery in
microspheres for oral delivery – effect of polysaccharide reinforce- rats by nanoparticles prepared with non-toxic solvents. Int J Pharm
ment on physicochemical properties and release profile. Carbohydr 443:169–74.
Polym 69:725–731. Wang XQ, Zhang, Q. (2012). pH-sensitive polymeric nanoparticles to
Minimol PF, Paul W, Sharma CP. (2013). PEGylated starch acetate improve oral bioavailability of peptide/protein drugs and poorly water-
nanoparticles and its potential use for oral insulin delivery. Carbohydr soluble drugs. Eur J Pharm Biopharm 82:219–29.
Polym 95:1–8. Werle M, Loretz B, Entstrasser D, et al. (2007). Design and evaluation of
Mueller C, Verroken A, Iqbal J, et al. (2011). Thiolated chitosans: a chitosan-aprotinin conjugate for the peroral delivery of therapeutic
in vitro comparison of mucoadhesive properties. J Appl Polym Sci peptides and proteins susceptible to enzymatic degradation. J Drug
126:5046–55. Target 15:327–33.
Mukhopadhyay P, Sarkar K, Chakraborty M, et al. (2013). Oral insulin Woitiski CB, Sarmento B, Carvalho RA, et al. (2011). Facilitated
delivery by self-assembled chitosan nanoparticles: in vitro and in vivo nanoscale delivery of insulin across intestinal membrane models. Int J
studies in diabetic animal model. Mater Sci Eng C 33:376–82. Pharm 412:123–31.
Niu M, Lu Y, Hovgaard L, et al. (2011). Liposomes containing Wong TW. (2010). Design of oral insulin delivery systems. J Drug Target
glycocholate as potential oral insulin delivery systems: preparation, 18:79–92.
in vitro characterization, and improved protection against enzymatic Wu ZM, Guo XD, Zhang LJ, et al. (2012a). Solvent mediated
degradation. Int J Nanomed 6:1155–66. microstructures and release behavior of insulin from pH-sensitive
Niu M, Lu Y, Hovgaard L, et al. (2012). Hypoglycemic activity and oral nanoparticles. Colloids Surf B 94:206–12.
bioavailability of insulin-loaded liposomes containing bile salts in Wu ZM, Ling L, Zhou LY, et al. (2012b). Novel preparation of
rats: the effect of cholate type, particle size and administered dose. PLGA/HP55 nanoparticles for oral insulin delivery. Nanoscale Res
Eur J Pharm Biopharm 81:265–72. Lett 7:299. doi:10.1186/1556-276X-7-299.
10 Y. Y. Luo et al. Drug Deliv, Early Online: 1–10

Wu ZM, Zhou L, Guo XD, et al. (2012c). HP55-coated capsule Zhang N, Ping QN, Huang GH, et al. (2005). Investigation of lectin-
containing PLGA/RS nanoparticles for oral delivery of insulin. Int J modified insulin liposomes as carriers for oral administration. Int J
Pharm 425:1–8. Pharm 294:247–59.
Xiong XY, Li QH, Li YP, et al. (2013). Pluronic P85/poly(lactic acid) Zhang X, Ma GH, Su ZG, et al. (2011a). Novel poly(L-lysine) particles
vesicles as novel carrier for oral insulin delivery. Colloids Surf B 111: for gene delivery. J Control Release 152:184–6.
282–8. Zhang X, Qi J, Lu Y, et al. (2014). Biotinylated liposomes as potential
Xiong XY, Li YP, Li ZL, et al. (2007). Vesicles from pluronic/poly(lactic carriers for the oral delivery of insulin. Nanomedicine 10:167–76.
acid) block copolymers as new carriers for oral insulin delivery. J Zhang X, Sun M, Zheng A, et al. (2012). Preparation and characteriza-
Control Release 120:11–17. tion of insulin-loaded bioadhesive PLGA nanoparticles for oral
Xiong XY, Tam KC, Gan LH. (2006). Polymeric nanostructures for drug administration. Eur J Pharm Biopharm 45:632–8.
delivery applications based on pluronic copolymer systems. J Nanosci Zhang Y, Wei W, Lv P, et al. (2011b). Preparation and evaluation of
Nanotechnol 6:2638–50. alginate-chitosan microspheres for oral delivery of insulin. Eur J
Yang J, Sun H, Song C. (2012). Preparation, characterization and Pharm Biopharm 77:11–19.
in vivo evaluation of pH-sensitive oral insulin-loaded poly(lac- Zhang Z, Shan H, Chen L, et al. (2013). Synthesis of pH-responsive
tic-co-glycolicacid) nanoparticles. Diabetes Obes Metab 14: starch nanoparticles grafted poly(l-glutamic acid) for insulin con-
358–64. trolled release. Eur Polym J 49:2082–91.
Yin L, Ding J, He C, et al. (2009). Drug permeability and mucoadhesion Zhao X, Shan C, Zu Y, et al. (2013). Preparation, characterization, and
properties of thiolated trimethyl chitosan nanoparticles in oral insulin evaluation in vivo of Ins-SiO(2)-HP55 (insulin-loaded silica coating
delivery. Biomaterials 30:5691–700. HP55) for oral delivery of insulin. Int J Pharm 454:278–84.
Yun Y, Cho YW, Park K. (2013). Nanoparticles for oral delivery: Zhu S, Qian F, Zhang Y, et al. (2007). Synthesis and characterization of
targeted nanoparticles with peptidic ligands for oral protein delivery. PEG modified N-trimethylaminoethylmethacrylate chitosan nanopar-
Adv Drug Deliv Rev 65:822–32. ticles. Eur Polym J 43:2244–53.
Downloaded by [Xiang Yuan Xiong] at 02:11 07 August 2015

View publication stats

You might also like