You are on page 1of 16

Send Orders for Reprints to reprints@benthamscience.

net

Pharmaceutical Nanotechnology, 2019, 7, 1-16 1


REVIEW ARTICLE

Nanotechnology in Insulin Delivery for Management of Diabetes

Fatemah Bahman, Khaled Greish* and Sebastien Taurin*

Department of Molecular Medicine, Princess Al-Jawhara Centre for Molecular Medicine, School of
Medicine and Medical sciences, Arabian Gulf University, Manama, Kingdom of Bahrain

Abstract: Diabetes is a group of diseases characterized by hyperglycemia and originating


from the deficiency of or resistance to insulin, or both. Ultimately, the most effective
treatment for patients with diabetes involves subcutaneous injections of insulin. However,
this route of administration is often painful and inconvenient, as most patients will have to
self-administer it at least twice a day for the rest of their lives. Also, infection, insulin pre-
ARTICLEHISTORY
cipitation, and either lipoatrophy or lipohypertrophy are frequently observed at the site of
injection. To date, several alternative routes of insulin administration have been explored,
Received: January 26, 2019 including nasal, pulmonary and oral. Although oral delivery of insulin is an ideal route for
Revised: February 22, 2019
Accepted: March 18, 2019 diabetic patients, there are several limitations that have to be overcome such as the rapid
DOI:
loss of insulin in the gastric fluid and low oral bioavailability. Numerous strategies have
10.2174/2211738507666190321110721 been carried out to improve these limited parameters such as the use of enzyme inhibitors,
absorption enhancers, mucoadhesive polymers and chemical modification for receptor-
mediated absorption. Also, insulin-loaded nanocarriers bypass several physiological barri-
ers. This current review focuses on the various barriers existing in the delivery of insulin
through the oral route and the strategies undertaken so far to overcome those obstacles us-
ing nanocarriers as a potential vehicle of insulin.

Keywords: Diabetes mellitus, insulin analog, insulin, nanoparticles, nasal delivery, oral delivery, pulmo-
nary delivery.

1. INTRODUCTION producing β-cells in the pancreas [1, 3]. T2DM is a


non-insulin dependent DM and has been associated
Diabetes mellitus (DM) is a metabolic syn-
to a combination of genetic, physiologic and envi-
drome regrouping a plethora of chronic patholo-
ronmental factors such as the age, obesity, and the
gies characterized by hyperglycemia and denoting
lack of physical activities [4, 5]. TD2M arises
the insufficient secretion and or action of insulin
from the peripheral resistance to insulin, leading to
[1]. DM is mainly classified into type I (T1DM)
insulin overproduction by β-cells and or a relative
and II (T2DM). Other types of DM, including ges-
insulin deficiency due to impaired β-cell function
tational diabetes and secondary forms of diabetes,
[6]. The global prevalence in adults has been in-
are less common [2]. T1DM is an autoimmune
creasing over the last decades as 8.8% of the adult
condition mediated by the destruction of insulin-
population worldwide has developed DM [7].
T1DM represents 7-12% of the DM cases world-
*Address correspondence to these authors at the College of wide [8], while T2DM is more common and has
Medicine and Medical Sciences, Department of Molecular increased by nearly 5-folds over the last 40 years
Medicine, and Nanomedicine Unit, Princess Al-Jawhara and is estimated to affect more than 500 million
Center for Molecular Medicine, Arabian Gulf University,
Manama, Kingdom of Bahrain; Tel: +973 17 237 393; and
people worldwide in 2018 [9, 10]. T2DM is ex-
+973 17 237; E-mails: khaledfg@agu.edu.bh; pected to further spread over the next few years
sebastient@agu.edu.bh among the middle and low-income counties as

2211-7385/19 $58.00+.00 © 2019 Bentham Science Publishers


2 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 Bahman et al.

more people adopt a sedentary lifestyle (Table 1) activated by the blood glucose level [19]. Some
[11, 12]. In 2016, the World Health Organization amino acids such as glutamine and leucine can in-
(WHO) estimated that DM was the 7th cause of directly influence β-cell insulin secretion. Leucine
death worldwide, 1.6 million deaths were directly can stimulate glutamate dehydrogenase, which
related to DM [13]. Furthermore, the total health converts glutamate into α-ketoglutarate. Gluta-
care budget associated with DM increased from mine, after being transformed into glutamate by
the US $246 in 2007 to $673 billion in 2015 [14] glutaminase and then α-ketoglutarate can enter the
and is estimated to rise to $2.2 trillion by 2030 citric acid cycle resulting in ATP production and
[15]. enhancement of insulin secretion [20]. Also, free
Table 1. Prevalence increased % of DM worldwide
amino acids including alanine and glutamine, are
in 2045 according to IDF 2018. released into the blood and served as signals to
glucagon secretion. This results in the elevation of
Prevalence blood glucose levels (BGLs), which then triggers
Area
Increased % insulin release into the circulation [21, 22].
Asia 15%
3. DIFFERENT TYPE OF RECOMBINANT
South East Asia 84% INSULIN
Middle East countries and North Africa 110%
The insulin used for the treatment of DM was
Africa 156% provided initially from porcine or cow pancreas
North America & Caribbean 35% extracts. However, the growing number of diabetic
patients and the concern over the development of
South and Central America 62%
allergic reaction led to the development of recom-
Europe 16% binant human insulin. In 1978, Genentech and City
of Hope National Medical Center produced the
2. INSULIN human insulin by inserting the genes into Esche-
richia coli [23]. The first recombinant human insu-
In 1921, F.G. Banting, C.H. Best, and J.J.R.
lin, Humulin, was approved by the FDA in 1982
Macleod were the first to discover insulin. In [24]. Recently, genetic engineering allowed the
1922, a 14-year-old patient with T1DM received
development of insulin analogs to provide the
the first injection of a pancreas extract, transform- better control of blood glucose. There are various
ing a deadly condition into a manageable disease
human insulin analogs in the market as shown in
[16]. The insulin hormone is a 51 amino acids pro- Table 2. Modifications made on the insulin back-
tein [17], forming an “A” (21 amino acids) and
bone to produce the different types of insulin ana-
“B” (30 amino acids) chain connected with a di- logs are shown in Fig. (1). Rapid-acting insulins,
sulfide bond. Insulin is synthesized into a single
such as Lispro [25], Aspart [26], and glulisine [27]
chain precursor called preproinsulin; the removal have a rapid onset of action of approximately 15-
of the signal peptide during insertion into the en-
30 min and a duration of action of about 4 to 5 h.
doplasmic reticulum creates proinsulin [18]. The Short-acting insulins such as Actrapid, Humulin,
proinsulin contains three domains, an amino-
Hypurin and Neutral [28] have an onset of action
terminal B chain, a carboxy-terminal A chain and of around 30 min to 1 h and a duration of action of
a connecting peptide called the C peptide. Within
approximately 6-8 h, with a peak at 2-4 h. Inter-
the endoplasmic reticulum (ER), the C peptide is
mediate-acting insulins such as Neutral Protamine
cleaved by endopeptidases, thereby producing the
Hagedorn (NPH) [29] and Lente [30] have an on-
mature form of insulin. Then, both insulin and free
set of action of 1-2 h, a peak action of 6-10 h and
C peptide are packaged in the Golgi as a micro-
approximately 10-16 h duration of action. Long-
crystalline zinc insulin hexamer into secretory
acting insulins such as an Insulin Detemir (desB30
granules which accumulate in the cytoplasm [19].
insulin) [26, 31], Glargine and Ultralente [32] have
The stimulation of β-cell leads to the exocytosis of
an onset of activity of 2 h, a peak of action of
insulin and diffusion into the islet blood capillaries
about 6-20 h and a duration of around 36 h.
[18]. Physiologically, the secretion of insulin is
Nanotechnology in Insulin Delivery for Management of Diabetes Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 3

Fig. (1). Amino acid sequence of human insulin and its analogs. a: Insulin lispro, the sequence changes from proline
B28 and lysine B29 to Lysine B28 and proline B29. b: Insulin glargine, sequence addition of arginine B31 and ar-
ginine B32, plus substitution alanine A21 to glycine. c: insulin aspart, substitution of proline B28 to aspartate B28
d: Insulin glulisine, substitutions of asparagine B3 to lysine B3 and lysine B29 to glutamate B29. e: Insulin detemir.
Acylation of lysine B29 with C14 fatty acid chain, and removal of threonine B30. Adapted from [33].

Table 2. Human insulin analogs on the market.

Onset (min-hrs) Duration (hrs) Chemical Structure

Rapid-acting insulin
Insulin Lispro 28(B)-L-lysine-29(B)-L-proline-human insulin
Insulin Aspart 15 - 30 min 4h-5h AspB28
Insulin glulisine 3(B)-Lys, 29(B)-Glu-human insulin
Short-acting insulin
Actrapid
Humulin
0.5 - 1 h 6h-8h
Hypurin
Neutral
(Table 2) contd…
4 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 Bahman et al.

Onset (min-hrs) Duration (hrs) Chemical Structure

Intermediate-acting insulin
Proline at position 29 in the B-chain is replaced
Neutral Protamine Hagedorn (NPH)
1 -2 h 10 h - 16 h by Lysine.
Lente
Long-acting insulin
Insulin detemir LysB29(Nε-tetradecanoyl) des(B30)
Insulin glargine 2h 36 h Gly A21, Arg B31, Arg B32
Insulin Ultralente

4. DIFFERENT STRATEGIES FOR INSULIN advantages of this method include that the precise
DELIVERY insulin dosages can be administered and calculat-
ed. However, the disadvantages include mechani-
Subcutaneous insulin delivery is currently the cal failure and inflammation at the injection site
preferred method of insulin delivery [34, 35]. Nu- [40]. Usage of the insulin pump is generally for
merous alternative strategies have been investigat- adults suffering from T1DM [41].
ed for insulin delivery than subcutaneous route
including oral, rectal, ocular, buccal, intravaginal, 4.2. Nasal Insulin
transdermal, pulmonary and nasal [36]. The intra-
muscular route (IM) of administration is also not a Intranasal insulin delivery has advantages be-
reliable route as the IM delivery route cannot cause the nasal cavity is the largest absorptive sur-
mimic the regular secretion pattern of insulin and face area in the body accounting for approximately
leads to change the pharmacokinetic and pharma- 150 cm2. Also, the mucosa is highly vascularized
codynamic properties of insulin [37]. The IM insu- and may promote the direct protein transport into
lin injections could lead to hypoglycemia immedi- the systemic circulation bypassing the first-pass
ately after injection and hyperglycemia before the metabolism by the liver and intestine [42]. How-
next dose [38]. ever, several factors may delay the intranasal ab-
sorption such as nasal cavity clearance, infiltration
4.1. Subcutaneous Insulin Injections across the mucus coating and enzymatic degrada-
tion similar to oral insulin delivery [43]. Intranasal
Subcutaneous injections called preprandial in- delivery of insulin has been reported by using li-
jections of rapid-acting insulin and predormital pid-based particles including liposomes, micro-
injections of long-acting insulin are the regimens emulsions and solid lipid nanoparticles (SLNs).
for patients with T1DM to maintain BGL when the Furthermore, insulin-loaded liposomes showed an
preprandial blood glucose is 3.9 to 7.2 mmol/L (70 effective absorption enhancer for the nasal insulin
to 130 mg/dl) and the 2 h postprandial blood glu- in rabbits and reduced BGL for 8 h with an insulin
cose less than 10 mmol/L (<180 mg/dl) [1]. The bioavailability of more than 13% [44, 45].
insulin is administered directly to the fatty layer
which is less vascularized and therefore slows the 4.3. Pulmonary Insulin
absorption rate. However, non-adherence and poor Insulin is usually delivered to the lungs in pow-
compliance are associated with insulin subcutane- der form and is delivered to the lungs with the help
ous injections [39]. of an inhaler. The mode of delivery is comparable
Insulin pumps usually work by delivering insu- but not superior to the traditional subcutaneous
lin continuously through the subcutaneous route. injectable route [34]. It is documented that inhaled
The pump mimics the body basal insulin release in insulin can also achieve a normal BGL of 70 to
24 h cycle. Bolus insulin can be administered to 130 mg/dl [46]. The lungs are well perfused hence
correct the high glycemic levels after meals. The increasing the surface area for absorption. Some of
Nanotechnology in Insulin Delivery for Management of Diabetes Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 5

the disadvantages are associated with the inhaled acidic condition. The gastric environment is char-
insulin that can precipitate acute episodes of bron- acterized by a harsh environment caused by
chospasm in asthmatic patients and may also cause peptidase activities and the acidic pH [59, 60].
hypoglycemic episodes and throat irritation [47]. One study showed that HP55 modification, one
Liposomes loaded with insulin enhance the pul- kind of hydroxypropyl methylcellulose phthalate,
monary uptake of insulin and improve hypogly- an enteric coating, protected insulin against acidic
cemic effect when compared with the free insulin gastrointestinal conditions and then dissolved in
[48]. Insulin-encapsulated in PLGA microcapsules the small intestine environment. This modification
demonstrated a constant insulin release with a reduced the cumulative release rate of insulin in
longer period of 4 to 5 fold longer than respirable simulated gastric fluid from 33.24 to 15.87%,
spray dried insulin powder, as well as a stable gly- while cumulative release rates of insulin in simu-
cemic level up to 24 h after administration [49]. lated intestinal fluid increased from 15 to 58.06%
The Exubera® was the first inhaled product ap- [61, 62]. Also, different studies have confirmed
proved by the FDA in 2006. It is a dry powder that the use of digestive enzyme inhibitors can
product available in 1 mg and 3 mg doses that can overcome this challenge such as sodium glycocho-
be taken with the help of an Inhance™ inhaler de-
late, bacitracin, soybean trypsin inhibitors, apro-
vice. The pharmacokinetic and pharmacodynamic
tinin, and camostat mesilate [63]. One approach
(PK/PD) properties were similar to the insulin As-
part with a 10-15 min onset of action [50]. In clin- uses different types of protease inhibitors such
ical trials with uncontrolled T1DM and T2DM pa- aprotinin, soybean trypsin inhibitors, and camostat
tients, Exubera showed a significant reduction in mesilate may result in the deficiency of these en-
postprandial BGL and Hb A1c [51]. Exubera was zymes in humans if they administrated for long
discontinued by Pfizer in 2007 due to its high period [64, 65]. One study found that tryp-
price, huge size and concerns over its effect on the sin/chymotrypsin inhibitor aprotinin was used to
lungs [52]. Afrezza is another dry powdered for- shield insulin and protect it from degradation [66].
mulation of inhaled insulin developed. Afrezza has Also, a new class of enzyme inhibitors called
an onset of action of 15 min, duration of 2-3 h, and chicken and duck ovomucoids has been docu-
was ideal for controlling postprandial BGL. There- mented, these formulations offered complete pro-
fore, it completed phase III clinical trials for pa- tection against the action of both enzymes trypsin
tients with T1DM and T2DM and was approved and chymotrypsin [67]. Moreover, polymer inhibi-
by the FDA in 2014 [53-55]. tor conjugates such as carboxymethyl cellulose-
elastin (CMC-Ela) have demonstrated protection
4.4. Oral Insulin against enzymes trypsin and elastase in vitro. After
Oral insulin administration is the most conven- 4 h of incubation, about 33% of the therapeutic
ient and ideal route for DM. However, the oral bi- protein was found to be active against the elastase
oavailability of insulin is usually less than 10%, [68]. Another approach to protect proteins is using
and it is reported that less than 0.5% of orally ad- Serpin (Serine protease inhibitor) that forms cova-
ministered insulin enters the systemic circulation lent complexes with the protease [69].
[56]. Also, there are many challenges associated
the oral method delivery. Some of these challenges 6. DIFFERENT FORMULATIONS FOR
include the activity of the proteolytic enzymes in ORAL INSULIN NANOPARTICLES
the GIT [57] and the poor bioavailability of oral Nanoparticles provide potent tools for the oral
insulin due to the low permeability of the intestine delivery of insulin such as 1) insulin encapsulation
[58]. within nanoparticle carriers protect it from acidic
pH and enzymatic degradation in the gastrointesti-
5. OVERCOMING PHYSIOLOGICAL BAR- nal tract (GIT); 2) the mucoadhesive coating of the
RIERS AND ENZYMATIC BARRIERS FOR nanoparticles increases the efficient drug delivery
ORAL DELIVERY near the site of absorption in the intestine; 3)
New studies geared towards changing the struc- mucus-penetrating particles or coated insulin na-
ture of the insulin analogs to be more stable in noparticle may improve transportation of insulin
6 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 Bahman et al.

and 4) stabilization of the poorly soluble insulin by persisted up to 10 h in diabetic rats [78]. Cui et al,
encapsulation into nanoparticle such as PLGA [57, formulated an insulin pH sensitive carboxylated
70]. chitosan grafted poly(methyl methacrylate) nano-
particles that lowered BGL after oral administra-
6.1. Nano-Medicine and Nano-Particles (Nano- tion at 50 and 25 IU/kg and the pharmacological
Carriers) that used for Insulin bioavailability was 9.7% at 25 IU/kg, 4.4% and
Recently, nanoparticles were developed to 3.2% at 50 and 100 IU/kg respectively, in diabetic
promote the oral delivery of insulin. The nanopar- rats [79].
ticles were either nanospheres where the insulin is Elsayed et al., prepared insulin-chitosan com-
uniformly distributed within the polymeric matrix plexed with oleic acid, plurol oleique as cosurfac-
or nanocapsules where the insulin is within the tant and labrasol as a surfactant and showed a sig-
membrane polymers (polymeric film) [71-73]. Ta- nificant hypoglycemic effect in diabetic rats after
ble 3 presents some of these nanoparticles used for oral insulin dose of 50 IU/kg throughout 24 h [80].
the oral delivery of insulin.
6.1.1.2. Alginate
6.1.1. Natural Polymers
Alginate is a nontoxic and biodegradable nano-
Polysaccharides (chitosan, dextran, and algi- carrier obtained from the marine brown algae. It is
nate) and proteins (casein and gelatin) are the most composed of 1,4-linked-β-D-mannuronic acid and
known naturally occurring polymers used in the α-L-guluronic acid residues that form polyelectro-
oral delivery of insulin. They are hydrophilic, non- lyte complexes with polycations such as chitosan.
toxic, biocompatible and biodegradable polymers.
Insulin-loaded nanoparticles were prepared by
6.1.1.1. Chitosan-Insulin Nanoparticles the ionotropic pre-gelation of alginate with calci-
Chitosan is a type of a linear cationic non-toxic um chloride followed by complexation between
polysaccharide, that adheres to the mucosal sur- alginate and chitosan and led to the formation of
face, hence causing the opening up of the tight epi- nanoparticles with a size of 748 nm [81]. The
thelial junctions [74]. Chitosan has a strong elec- pharmacological effect of these nanoparticles was
trostatic affinity towards mucin, is the only natural evaluated in diabetic rats at 50 and 100 IU/kg dos-
polysaccharide with good biodegradability, bio- es and showed a significant reduction in BGL by
compatibility, and has an excellent adhesion prop- more than 40% with 50 IU/kg and 100 IU/kg dos-
erty [75]. The encapsulation of insulin in chi- es, while the hypoglycemic effect was sustained
tosan/heparin nanoparticles demonstrated a normal over 18 h [82].
glycemic control [76]. This study used a dose of The alginate/chitosan nanoparticles that form
60 IU/kg that lowered the mean BGL by 30% for 2 complexes with cationic β-cyclodextrin polymers
h after insulin administration and lasted for 4 h; will be able to protect insulin against degradation
also it provided 50% lower BGL at 3 h after ad- in simulated gastric fluid [83]. Also, insulin-
ministration that lasted for 5 h [76]. Another study loaded dextran sulfate/chitosan nanoparticles with
showed that chitosan nanoparticles encapsulating a size of 527 nm, when given orally at a dose of 50
insulin (CS-NPs) had a size between 265.3 ± 34 to and 100 IU/kg resulted in low BGL of 35%. The
387.4 ± 35.6 nm, and administration of 21 IU/kg rats experienced the hypoglycemic effects for over
resulted in the hypoglycemic effect that prolonged 18 h [82].
up to 15 h and an average pharmacological bioa-
vailability relative to subcutaneous injection of Ma et al. prepared a chitosan insulin nanoparti-
insulin up to 14.9% [77]. cle composed of chitosan, and TPP (pentasodium
tripolyphosphate) sized 269 to 688 nm, respective-
Lin et al. developed a nanoparticle of chitosan ly. These nanoparticles were effective in lowering
within Poly (γ-glutamic acid) sized between 110 to the BGL of diabetic rats when taken orally at insu-
150 nm that resulted in a hypoglycemic effect after lin doses of 50 and 100 IU/kg. In particular, the
oral administration of doses of 15 and 30 IU/kg, oral insulin dose of 100 IU/kg was able to main-
which showed a significant decrease of BGL that tain the BGL for at least 11 h [84].
Nanotechnology in Insulin Delivery for Management of Diabetes Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 7

Table 3. Different types of oral insulin nanocarrier.

Route of Dose (I.U./kg)


Nano-Carrier Components Size (nm)
Administration (min/max)

Carboxylated chitosan + methyl


251 to 319 Oral 15 (min)- 100 (max)
methacrylate
Polysaccharide chitosan nanoparticules 265.3 ± 34 to
Oral 21 (max)
(CS-NPs) 387.4 ± 35.6
Chitosan + Oleic acid + Plurol oleique +
108 Oral 50
Labrasol
25 (min)
Chitosan + Alginate 748 Oral
100 (max)
50 (min)
Chitosan + Dextran sulfate 527 Oral
100 (max)

Chitosan-insulin Chitosan + TPP (pentasodium 7 (min)


250 to 400 Oral
tripolyphosphate) + Poloxamer 188 21 (max)
50 (min)
Chitosan + TPP(pentasodium
269 to 688 Oral
tripolyphosphate) 100 (max)

15 (min)
Chitosan + Poly(ç-glutamic acid) 110 to 150 Oral
30 (max)
Chitosan + Alginate + Calcium chloride + 25 (min)
Labrafac CC + Phospholipid + Span 80 + 488 Oral
Cremorphor EL 50 (max)

Chitosan + γ-PGA 185.1 to 198.4 Oral 30


PLGA + Phospholipid + PVA 102 to 428 Oral 20
PLGA + Hp55 169 Oral 20
PLGA + Chitosan + Pluronic 188 134.4 Oral 15
PLGA-Insulin
PLGA + Pluronic 188 121.3 Oral 15
PLGA + Sodium oleate + PVA 161 Oral 20
PLGA + PEG + Folate ∼260 Oral 50
25 (min)
PCL-Insulin PCL and Eudragit® RS 331 Oral
100(max)
Dextran + Epichlorohydrin + vitamin
160 to 250 Oral 20
B(12)
Dextran + Alginate + Poloxamer + Chito-
Dextran- Insulin 396 Oral 50
san + BSA

Dextran + Alginate + Chitosan + PEG + >1842 (90 %) 25 (min)


Oral
BSA >812 (50 %) 100 (max)
(Table 3) contd…
8 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 Bahman et al.

Route of Dose (I.U./kg)


Nano-Carrier Components Size (nm)
Administration (min/max)

Isopropyl myristate + Labrasol + Plurol


200 to 400 Oral 100
Oleique + butyl (2) cyanoacrylate

Polyalkylcy- Polybutylcyanoacrylate + Tween 20 78 Oral 50


anoacrylate-insulin Polybutylcyanoacrylate + Tween 20 +
67 Oral 50
Soyabean oil + vitamin E
Isobutyl cyanoacrylate (IBCA)+ insulin 145 Oral 100
Lecithin + stearic acid + ploxamer + wheat
germ agglutinin-N-glutamyl-phosphatidyl- 75.3 Oral 50
ethanolamine
Stearic acid/octadecyl alcohol/cetyl palmi-
Solid lipid-insulin tate/glyceryl monostearate/glyceryl palmi- 213 to 444.8 Oral 50
tostearate/glyceryl tripalmitate/glyceryl
behenate
Witepsol 85E 243 ± 10 Oral 25
Witepsol 85E + chitosan 470 ± 32 Oral 25
N-trimethyl chitosan chloride +
Targeted insulin 342 Oral 50
CSKSSDYQC peptide +
nanoparticle
PLA-PEG + human polyclonal IgG Fc 63 Oral 1.1

Li, X et al. prepared a nanoparticle by prepar- teases [86]. Dextran is normally conjugated to vit-
ing an insulin solution composed of chitosan, algi- amin B-12, then when this complex reaches the
nate and calcium chloride supplemented to the oily intestines, it escapes the obstacles in the digestion
phase which was composed of 68.5% Labrafac system [72, 87].
CC, 25% Span™ 80, and 6.5% phospholipid; Then
Chalasani et al. developed a VB12 nanoparticle
this emulsion was kept into an aqueous solution
system that improves the uptake capacity of both
containing 3% Cremophor EL (castor oil deriva-
nanoparticles and VB12 transport to deliver insu-
tive) and low-viscosity sodium alginate and when
lin orally. The results of this study concluded that
administered orally at the insulin doses of 12.5
20 IU/kg of insulin conjugated with VB12-
IU/kg, 25 IU/ kg, and 50 IU/kg, it resulted in lower
Dextran NPs showed a significant BGL reduction
BGL and pharmacological bioavailability of 25
up to 24 h [87].
and 50 IU/kg was 8.19% ± 0.58% and 7.84% ±
0.29%, respectively [85]. Lin Y et al,. prepared a Reis et al. created an oral insulin nanoparticle
nanoparticle composed of chitosan and poly (ç- based on alginate‐dextran sulfate core, capsulated
glutamic acid) sized 185.1 to 198.4 nm that with a chitosan‐polyethylene glycol albumin shell.
showed a reduction in BGL at a dose of 30 IU/kg Albumin was supplemented to avoid protease ac-
and hypoglycemic effects were present for up to tion on the destruction of insulin and chitosan be-
12 h [78]. cause of mucoadhesive properties, while PEG
helped the stabilizer to improve the insulin half-
6.1.1.3. Dextran-Insulin Nanoparticles
life. Chitosan-PEG-albumin nanoparticle mini-
The dextran sulfate is an exocellular bacterial mized the BGL up to 70%. Therefore, the insulin‐
polysaccharide that is a nontoxic, highly water- loaded chitosan-PEG-albumin nanoparticle with
soluble, biodegradable and biocompatible poly- 25, 50, 100 IU/kg administered orally to diabetic
mer. Dextran is chemically coupled to insulin and rats caused a reduction in BGL, lasted for 24 h
acts by protecting the insulin against the GIT pro- with a maximal effect after 14 h [88].
Nanotechnology in Insulin Delivery for Management of Diabetes Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 9

Woitiski et al. produced a multilayer nanoparti- Also, the same group prepared PLGA nanopar-
cle sized 396 nm that consisted of alginate and ticles (PNP) and PLGA-Hp55 (PHNP) nanoparti-
dextran sulfate nucleating around calcium and cles sized 150 ± 17 and 169 ± 16 nm, respectively,
binding to poloxamer, stabilized by chitosan, and and resulted in an effective reduction in BGL after
coated with albumin. This nanoparticle adminis- oral administration [95]. Zhang et al. developed a
tered orally to diabetic rats (50 IU/kg) reduced mucoadhesive PLGA nanoparticle coated with chi-
BGL to 40% and continued hypoglycemic effect tosan or Eudragit® RS to enhance the oral bioa-
for over 24 h [89]. vailability. These nanoparticles were prepared by
water-in oil-in-water solvent evaporation tech-
6.1.2. Synthetic Polymers nique; insulin was dissolved in an aqueous solu-
tion of Pluronic 188, PLGA and an aqueous solu-
Poly ɛ-caprolactone (PCL), poly (lactic-co-
tion of Pluronic 188, to form a double emulsion
glycolic acid) (PLGA) and polylactides (PLA) are
(w/o/w). This preparation was tested orally on dia-
synthetic polymers and were used in oral insulin
betic animals which resulted in hypoglycemia, and
delivery due to their good biocompatibility and
pharmacological availability of both nanoparticles,
biodegradability.
PLGA-NP and CS-PLGA-NP, relative to the sub-
6.1.2.1. Poly (Lactic-co-Glycolic Acid) (PLGA) cutaneous injection was calculated to be 7.6% and
10.5%, respectively, at the dose of 15 IU/kg [96].
Poly (lactic-co-glycolic acid) (PLGA) is an ali- Wu Z et al. concluded that the pharmacological
phatic polyester synthetic biodegradable biopoly- availability of the 50 IU/kg Eudragit® RS (RS)
mer. Yang et al. prepared PLGA nanoparticles coated PLGA nanoparticle was 9.2% [97].
loaded with insulin. The administration of insulin-
loaded PLGA nanoparticles induced a rapid drop Sun et al. prepared an insulin-S.O (Ins-S.O) co-
in BGL for up to 24 h [90]. PLGA has the ad- mplex by a hydrophobic ion pairing (HIP) method
vantage of passing through the GIT and be ab- of size 161 nm. These nanoparticles reduced BGL
sorbed by the Peyer’s patches by the M cells in the to 23.85% for up to 12 h post-oral administration
intestine. PLGA nanoparticles have been exten- and the effect was prolonged for 24 h [98].
sively used because of their encapsulating proper- Jain S et al. showed that the oral administration
ties and their ability to release a drug in a con- of (50 IU/kg) of insulin encapsulated in folate-
trolled way [49]. Although there is a prolonged (FA) coupled with polyethylene glycol (PEG)
release with the use of PLGA nanoparticles, it has ylated polylactide-co-glycolide (PLGA) nanoparti-
been challenging how to monitor the amount of cles (NPs; FA-PEG-PLGA NPs) exhibited a hypo-
drug released per day and compromise the glyce- glycemic effect for 24 h [99].
mic control [91]. PLGA has shown good bioavail-
ability when administered with chitosan [92]. 6.1.2.2. Polylactide (PLA)
PLGA contributes to the stability of the colloidal PLA has similar characteristics to PLGA but
system and its transport across the mucosal surfac- they are hydrophobic, and they degrade more
es [93]. slowly because of the crystallinity (100). As men-
Cui F et al. prepared a novel nanoparticle com- tioned before, Cui et al. reported enhanced insulin
posed of dichloromethane (DCM). Ethyl acetate efficiency (up to 90%) in PLA and PLGA nano-
(EAc) containing 2% polymer (w/v) was added to particles, where insulin was encapsulated with
the insulin -phospholipid (Ins-SPC) complex, and phosphatidylcholine (SPC) to improve the lipo-
poured into an aqueous solution containing 2% solubility and oral bioavailability of 7.7% [94].
Poly (vinyl alcohol), in which soybean phosphati- 6.1.2.3. Poly-ɛ caprolactone (PCL)
dylcholine (SPC) was used to improve the lipo-
solubility of insulin. This formulation showed that It is a biodegradable polyester polymer with a
oral administration of 20 IU/kg (Ins-SPC) nano- semi‐crystalline feature. It has superior viscoelastic
particles reduced the fasting BGL to 57.4% within properties and the advantage that it generates a
the first 8 h and this continued for 12 h [94]. less acidic environment during degradation as
10 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 Bahman et al.

compared with PLGA [100]. Damgé et al. group The main advantage of SLNs as nanocarrier sys-
developed a nanoparticle from a mixture of poly- tems is their physiological lipid components that
ester poly (ε-caprolactone), and a polycationic reduce the risk of toxicity and protect the proteins
acrylic polymer (Eudragit® RS) sized 331nm. The- from degradation by GIT enzymes [106].
se carriers used for delivering insulin orally proved
Zhang et al. prepared lectin-modified SLN to
a significant prolonged hypoglycemic effect in
entrap insulin. These SLNs were further modified
both diabetic and normal rats [101].
with wheat germ agglutinin-N-glutaryl-phospha-
6.1.3. Polyalkylcyanoacrylate-Insulin Nanoparti- tidylethanolamine (WGA). Both formulations pro-
cles tected insulin from degradation by GIT enzymes.
Furthermore, a reduction in BGL following oral
Initially, it was used in tissue “glue” during gavage was observed in rats with both formula-
surgery. However, many studies have demonstrat- tions. The pharmacological bioavailability follow-
ed its importance in the delivery of insulin orally, ing oral administration of insulin-SLNs and WGA-
because of its stability and its biodegradable char-
modified insulin-SLNs was 4.46% and 6.08%, re-
acter [86]. It is a polymeric colloidal drug carrier,
spectively [107].
with isobutyl cyanoacrylate nanocapsules with less
than 300 nm in diameter. These nanoparticles can Fonte et al. developed chitosans covered with
pass the intestinal epithelium cells and protect in- Witepsol 85E solid lipid nanoparticles (SLN) to
sulin from proteolytic degradation [102]. Also, it encapsulate insulin. The diameters of SLN and
was designed by interfacial emulsion polymeriza- chitosan-SLN were 243 ± 10 nm and 470 ± 32 nm,
tion of a cyanoacrylic monomer where the oil respectively. The oral administration of chitosan-
phase (Miglyol 812) containing insulin was mixed coated insulin SLN to diabetic rats decreased the
to an aqueous medium in the presence of polox- BGL up to 24 h [108].
amer 188 (surfactant agent). Damgé et al. group
prepared insulin nanospheres composed of a pol- Yang et al. group created a solid lipid nanopar-
ymeric carrier formed by polymerization of isobu- ticle composed of stearic acid, octadecyl alcohol,
tyl cyanoacrylate (IBCA) in an acidic pH of size cetyl palmitate, glyceryl monostearate, glyceryl
145 nm. These nanocapsules showed a hypogly- palmitostearate, glyceryl tripalmitate and glyceryl
cemic effect after 2 h of nanosphere administration behenate of size about 213 to 444.8 nm. This for-
of 100 IU/kg [102]. mulation showed a hypoglycemic effect that lasted
for 24 h after oral administration of 50 IU/kg
Hou et al. group prepared two formulations of [109].
insulin-polybutylcyanoacrylate nanoparticle (IPN)
in soybean oil containing 0.5% (v/v) Tween-20 More recently, Xu et al. developed an insulin-
and 5% (v/v) Vitamin E (size 67 nm) and poly- loaded SLN with endosomal escape peptide. The
butylcyanoacrylate containing 0.5% (v/v) Tween- oral administration of this insulin-loaded SLN (50
20 (size 78 nm), which showed the hypoglycemic IU/kg) transiently decreased BGL in diabetic by
effect following oral administration of (50 IU/kg) nearly 35% after 3 h which was still low by almost
to diabetic rats [103]. 20% after 12 h. In comparison, insulin (5 IU/kg)
given subcutaneously decreased BGL by 70% af-
Graf et al. group prepared a nanoparticle of in- ter 2 h but returned to its original level after 12 h
sulin in poly(alkyl cyanoacrylate) nanoparticles [110].
composed of isopropyl myristate, caprylocaproyl
macrogolglycerides, polyglyceryl oleate and insu- 6.1.5. Targeted Insulin Nanoparticles
lin of size 200-400nm that showed a hypoglycemic
Jin et al. developed a goblet cell-targeting na-
effect for up to 36 h after oral administration of
noparticle that consisted of N-trimethyl chitosan
100 IU/Kg [104].
chloride (TMC) modified with a CSKSSDYQC
6.1.4. Solid Lipid-Insulin Nanoparticles (SLN) peptide (CSK) targeting insulin. The results of in
vivo studies on diabetic animals showed better hy-
In 1990, colloidal nanocarriers prepared from poglycemic effect 1.5 fold compared to the un-
solid lipids have been used as nanoparticles [105]. modified formulation [111].
Nanotechnology in Insulin Delivery for Management of Diabetes Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 11

Pridgen et al. prepared PLA-PEG, and human calcium phosphate enteric-coated capsules. The
polyclonal IgG Fc targeted to the neonatal Fc re- formulation was tested in phase I clinical trial
ceptor (FcRn) of size 63 nm. This nanoparticle [118]. Macrulin is a lecithin-based microemulsion
showed a significant hypoglycemic effect in dia- developed to enhance the bioavailability of insulin
betic mice after oral administration [112]. [119]. Macrulin is currently in phase II clinical
trial in Europe for the treatment of T1DM and
7. ORAL INSULIN NANOFORMULATIONS T2DM. Jordanian Pharmaceutical Manufacturing
IN CLINICAL TRIALS Co has developed an oral liquid insulin delivery
system in which insulin-loaded chitosan nanopar-
Over the last few years, laboratories have de- ticles are dispersed in an oily vehicle [120]. The
veloped various nanoformulations to improve the preparation was assessed in phase I clinical trial
oral delivery of insulin. However, the biocompati- and demonstrated promising bioavailability [121].
bility of these nanocarriers, low oral bioavailabil- Novo Nordisk A/S developed several insulin for-
ity and large interindividual variations in insulin mulations utilizing Merrion’s GIPET® Technology
absorption have limited their transition to the clin- consisting of a lipid mixture or lipid microemul-
ic. Few oral insulin formulations developed by sion with a surfactant in an enteric-coated gel cap-
pharmaceutical companies are now being assessed sule [122]. The various formulations were tested in
in clinical trials (Table 4) [113, 114]. Oral-lyn is phase I clinical trials for patients with T2DM (Ta-
an insulin spray, developed by Generex Biotech- ble 4). The assessment of NN1952 ceased due to
nology, that has been approved in Ecuador and the interaction with the food intake [123]. Oshadi
Lebanon but still in phase III clinical trials in Eu- Drug Administration Ltd. has developed Icp-
rope, Canada, and the USA. Oral-Lyn was granted insulin, proinsulin, and C-peptide in Oshadi carrier
approval by the FDA under the Investigational for the oral delivery of insulin [115]. The Oshadi
New Drug (IND) program for the treatment of carrier is composed of pharmacologically inert sil-
T1DM and T2DM patients with severe or life- ica nanoparticles with a hydrophobic surface and
threatening conditions [115]. The Oral-lyn was branched polysaccharide [115]. The formulation was
developed for delivery of insulin via the buccal assessed in phase I and II clinical trials (Table 4).
mucosa to bypass the liver and GI tract. The for-
mulation consists of insulin-loaded micelle with a In addition to the new era of nanomedicine to
size above 7 µm and a surfactant to enhance ab- treat diabetes, few alternative insulin delivery sys-
sorption [115, 116]. The other formulations devel- tems are being developed such as a glucose-
oped for oral delivery of insulin are still in early responsive closed-loop insulin delivery system via
phases I or II. IN-105 developed by Biocon is a a painless microneedle array patch that contains
modified insulin conjugated through a spacer to a insulin encapsulated vesicles sensitive to both hy-
small polyethylene glycol molecule which im- poxia and hydrogen peroxide [124]. Another sys-
proves its stability. The drug is currently in phase tem conjugated glucosamine and insulin (Glc- in-
II clinical trial [117]. Diasome Pharmaceutical has sulin) to promote binding on red blood cell mem-
developed an insulin-loaded liposome harboring branes. The binding is reversible and releases insu-
on its surface a biotin-phosphatidylethanolamine lin in the setting of hyperglycemia [125].
to target the liver [115]. The insulin-loaded
hepatocyte-directed vesicle (HDV-I) has a diame- CONCLUSION
ter below 150 nm and is stable in the blood and the Oral insulin administration constitutes a tre-
upper GI. The low amount of insulin in each cap- mendous challenge to the management of DM and
sule (5U) and the targeting to the hepatocytes are the improvement in patient compliance. Nanomed-
designed to maximize efficiency and decrease the icine-based drug delivery systems are designed to
risk of insulin overdosing [116]. HDV-I was as- overcome barriers including the enzymatic degra-
sessed in several clinical trials in phase II and one dation, low intestinal permeability and bioavaila-
in phase III for T1DM and T2DM (Table 4). NOD bility, which may contribute to overcome these
pharmaceuticals developed Nodlin which consists challenges. This review has presented several
of insulin nanoparticles embedded in bio-adhesive nanocarriers and strategies tested in vivo for the
12 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 Bahman et al.

Table 4. Lists of oral insulin formulation systems in different clinical phases.

S. No. Company Name Product Name Product Technology Action Development Phase

1 Generex Oral-Lyn ™ Micelle-Penetration enhancers Short III (NCT00668850)


Biocon/
2 IN-105 PEG Conjugated insulin Short II (NCT03430856)
Bristol-Myers Squibb
II (NCT02794155)
Diasome Pharmaceuticals, HDV-insulin Hepatic-directed vesicle-insulin II-III (NCT00814294)
3 Short
Inc (HDV-1) (liposomal nanocarrier) II (NCT03156361)
II (NCT03096392)
Insulin nanoparticles with
NOD Pharmaceuticals, I (ChiCTR-TRC-
4 Nodlin bioadhesive nanoencapsulation Intermediate
Inc./ Shanghai Biolaxy, Inc 12001872)
(NOD Tech)
5 Provalis PLC Macrulin ™ Lipid-based microemulsion Short II
Jordanian Pharmaceutical Liquid delivery system with
6 JPM oral insulin I
Manufacturing Co. PLC insulin-chitosan nanoparticles
Insulin analog with an oral delivery
7 Novo Nordisk A/S NN1952 Short I (NCT01028404)
system GIPET®
I (NCT01334034)
OI338GT Insulin analog with an oral delivery
8 Novo Nordisk A/S Long I (NCT01931137)
(NN1953) system GIPET®
I (NCT01796366)
OI362GT Insulin analog with an oral delivery
9 Novo Nordisk A/S Long I (NCT01597713)
(NN1954) system GIPET®
OI287GT Insulin analog with an oral delivery
10 Novo Nordisk A/S Long I (NCT01809184)
(NN1956) system GIPET®
I (NCT01120912)
Oshadi Drug Insulin, proinsulin, and C-peptide in
11 Oshadi Icp. Short II (NCT01973920)
Administration Ltd. Oshadi carrier
I-II (NCT01772251)

oral delivery of insulin to control the level of glu- REFERENCES


cose in the blood. [1] American Diabetes A. Diagnosis and classification of
diabetes mellitus. Diabetes Care. 2014; 37 Suppl 1:
CONSENT FOR PUBLICATION S81-90.
[2] Owens DR, Zinman B, Bolli GB. Insulins today and
Not applicable. beyond. Lancet. 2001; 358(9283): 739-46.
[3] Atkinson MA, Eisenbarth GS, Michels AW. Type 1
CONFLICT OF INTEREST diabetes. Lancet. 2014; 383(9911): 69-82.
[4] Nair M. Diabetes mellitus, part 1: physiology and
The authors declare no conflict of interest, fi- complications. Br J Nurs. 2007; 16(3): 184-8.
nancial or otherwise. [5] Kumar D, Mukherjee K. Economic impact of type-2
diabetes mellitus on households in Hisar district of
Haryana state, India. 2014; 2(4): 125-129.
ACKNOWLEDGEMENTS [6] Barrett T. Type 2 diabetes mellitus: incidence, man-
agement and prognosis. Paediatrics and Child Health.
Declared none. 2013; 23(4): 163-7.
Nanotechnology in Insulin Delivery for Management of Diabetes Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 13

[7] Cho NH, Shaw JE, Karuranga S, Huang Y, da Rocha [23] Chance RE, Frank BH. Research, development, pro-
Fernandes JD, Ohlrogge AW, et al. IDF Diabetes At- duction, and safety of biosynthetic human insulin.
las: Global estimates of diabetes prevalence for 2017 Diabetes care. 1993; 16(Supplement 3): 133-42.
and projections for 2045. Diabetes Res Clin Pract. [24] Sanchez-Garcia L, Martín L, Mangues R, Ferrer-
2018; 138: 271-81. Miralles N, Vázquez E, Villaverde A. Recombinant
[8] Ogurtsova K, da Rocha Fernandes JD, Huang Y, Lin- pharmaceuticals from microbial cells: a 2015 update.
nenkamp U, Guariguata L, Cho NH, et al. IDF Diabe- Microbial cell factories. 2016; 15(1): 33.
tes Atlas: Global estimates for the prevalence of dia- [25] Holleman F, Hoekstra JB. Insulin lispro. N Engl J
betes for 2015 and 2040. Diabetes Res Clin Pract. Med. 1997; 337(3): 176-83.
2017; 128: 40-50. [26] Raskin P, Guthrie RA, Leiter L, Riis A, Jovanovic L.
[9] Kaiser AB, Zhang N, Der Pluijm WV. Global Preva- Use of insulin aspart, a fast-acting insulin analog, as
lence of Type 2 Diabetes over the Next Ten Years the mealtime insulin in the management of patients
(2018-2028). Diabetes. 2018; 67(Supplement 1): 202- with type 1 diabetes. Diabetes Care. 2000; 23(5): 583-
LB. 8.
[10] Katsarou A, Gudbjornsdottir S, Rawshani A, Dabelea [27] Becker RH, Frick AD. Clinical pharmacokinetics and
D, Bonifacio E, Anderson BJ, et al. Type 1 diabetes pharmacodynamics of insulin glulisine. Clin Pharma-
mellitus. Nat Rev Dis Primers. 2017; 3: 17016. cokinet. 2008; 47(1): 7-20.
[11] Atlas Diabetes. "International diabetes federation." [28] Ahmad K. Insulin sources and types: a review of in-
IDF Diabetes Atlas, 7th edn. Brussels, Belgium: In- sulin in terms of its mode on diabetes mellitus. J
ternational Diabetes Federation (2015). Tradit Chin Med. 2014; 34(2): 234-7.
[12] Werfalli M, Musekiwa A, Engel ME, Ross I, Kengne [29] Li J, Kuang Y. Pharmacokinetical models of subcuta-
AP, Levitt NS. The prevalence of type 2 diabetes neous injection of insulin analogues for type 1 diabe-
mellitus among older people in Africa: a systematic tes. 2008.
review study protocol. BMJ Open. 2014; 4(6): [30] Deckert T. Intermediate-acting insulin preparations:
e004747. NPH and lente. Diabetes Care. 1980; 3(5): 623-6.
[13] Global report on diabetes. World Health Organiza- [31] Havelund S, Ribel U, Plum A, Jonassen I, Volund A,
tion. 2018: 1-88. Markussen J, et al. The mechanism of protraction of
[14] Zhang P, Gregg E. Global economic burden of diabe- insulin detemir, a long-acting, acylated analogue of
tes and its implications. Lancet Diabetes Endocrinol. human insulin. Diabetes. 2004; 53: A109.
2017; 5(6): 404-5. [32] Saltiel AR, Kahn CR. Insulin signalling and the regu-
[15] Bommer C, Sagalova V, Heesemann E, Manne- lation of glucose and lipid metabolism. Nature. 2001;
Goehler J, Atun R, Barnighausen T, et al. Global 414(6865): 799.
Economic Burden of Diabetes in Adults: Projections [33] Varewijck AJ, Janssen JA. Insulin and its analogues
From 2015 to 2030. Diabetes Care. 2018; 41(5): 963- and their affinities for the IGF1 receptor. Endocr
70. Relat Cancer. 2012; 19(5): F63-75.
[16] Rosenfeld L. Insulin: discovery and controversy. Clin [34] Shah RB, Patel M, Maahs DM, Shah VN. Insulin
Chem. 2002; 48(12): 2270-88. delivery methods: Past, present and future. Int J
[17] Al Rubeaan K, Rafiullah M, Jayavanth S. Oral insulin Pharm Investig. 2016; 6(1): 1-9.
delivery systems using chitosan-based formulation: a [35] Al-Tabakha MM, Arida AI. Recent challenges in in-
review. Expert Opin Drug Deliv. 2016; 13(2): 223-37. sulin delivery systems: a review. Indian J Pharm Sci.
[18] Weiss M, Steiner DF, Philipson LH. Insulin biosyn- 2008; 70(3): 278-86.
thesis, secretion, structure, and structure-activity rela- [36] Alai MS, Lin WJ, Pingale SS. Application of poly-
tionships. InEndotext [Internet] 2014 Feb 1. MDText. meric nanoparticles and micelles in insulin oral deliv-
com, Inc. ery. J Food Drug Anal. 2015; 23(3): 351-8.
[19] Fu Z, Gilbert ER, Liu D. Regulation of insulin syn- [37] Kalra S, Gupta Y. Clinical applications of intramus-
thesis and secretion and pancreatic Beta-cell dysfunc- cular insulin. Journal of Endocrinology and Metabo-
tion in diabetes. Curr Diabetes Rev. 2013; 9(1): 25- lism. 2014; 4(3): 40-3.
53. [38] Vaag A, Pedersen KD, Lauritzen M, Hildebrandt P,
[20] Sener A, Malaisse WJ. L-leucine and a nonmetabo- Beck-Nielsen H. Intramuscular versus subcutaneous
lized analogue activate pancreatic islet glutamate de- injection of unmodified insulin: consequences for
hydrogenase. Nature. 1980; 288(5787): 187-9. blood glucose control in patients with type 1 diabetes
[21] McClements DJ. Encapsulation, protection, and de- mellitus. Diabet Med. 1990; 7(4): 335-42.
livery of bioactive proteins and peptides using nano- [39] Kim H, Park H, Lee SJ. Effective method for drug
particle and microparticle systems: A review. Adv injection into subcutaneous tissue. Scientific Reports.
Colloid Interface Sci. 2018; 253: 1-22. 2017; 7(1): 9613.
[22] Tang-Christensen M, Larsen PJ, Thulesen J, Nielsen [40] McAdams BH, Rizvi AA. An overview of insulin
JR, Vrang N. Glucagon-like peptide 2, a neurotrans- pumps and glucose sensors for the generalist. Journal
mitter with a newly discovered role in the regulation of clinical medicine. 2016; 5(1): 5.
of food ingestion Ugeskr Laeger. 2001; 163(3): 287- [41] Heinemann L, Fleming GA, Petrie JR, Holl RW, Ber-
91. genstal RM, Peters AL. Insulin pump risks and bene-
14 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 Bahman et al.

fits: a clinical appraisal of pump safety standards, ad- ing complexation and pH-sensitive hydrogels as de-
verse event reporting and research needs. A joint livery vehicles. european journal of pharmaceutical
statement of the European Association for the Study sciences. 2006; 29(3-4): 183-97.
of Diabetes and the American Diabetes Association [57] Ansari M. Oral Delivery of Insulin for Treatment of
Diabetes Technology Working Group. Diabetologia. Diabetes: Classical Challenges and Current Opportu-
2015 May 1; 58(5): 862-70. nities. Journal of Medical Sciences. 2015; 15(5): 209-
[42] Hinchcliffe M, Illum L. Intranasal insulin delivery 20.
and therapy. Adv Drug Deliv Rev. 1999; 35(2-3): [58] Chellappan DK, Yenese Y, Wei CC, Chellian J, Gup-
199-234. ta G. Oral Insulin: Current Status, Challenges, and
[43] Costantino HR, Illum L, Brandt G, Johnson PH, Quay Future Perspectives. Journal of Environmental Pa-
SC. Intranasal delivery: physicochemical and thera- thology, Toxicology and Oncology. 2017; 36(4).
peutic aspects. Int J Pharm. 2007; 337(1-2): 1-24. [59] Ahmad N, Mohd Amin MCI, Ismail I, Buang F. En-
[44] Maitani Y, Takayama K, Nagai T. The effect of soy- hancement of oral insulin bioavailability: in vitro and
bean-derived sterol and its glucoside as an enhancer in vivo assessment of nanoporous stimuli-responsive
of nasal absorption of insulin in rabbits in vitro and in hydrogel microparticles. Expert opinion on drug de-
vivo. International journal of pharmaceutics. 1995; livery. 2016; 13(5): 621-32.
117(2): 129-37. [60] Muheem A, Shakeel F, Jahangir MA, Anwar M, Mal-
[45] Muramatsu K, Maitani Y, Takayama K, Nagai T. The lick N, Jain GK, et al. A review on the strategies for
relationship between the rigidity of the liposomal oral delivery of proteins and peptides and their clini-
membrane and the absorption of insulin after nasal cal perspectives. Saudi Pharm J. 2016; 24(4): 413-28.
administration of liposomes modified with an en- [61] Zhao X, Shan C, Zu Y, Zhang Y, Wang W, Wang K,
hancer containing insulin in rabbits. Drug develop- et al. Preparation, characterization, and evaluation in
ment and industrial pharmacy. 1999; 25(10): 1099- vivo of Ins-SiO2-HP55 (insulin-loaded silica coating
105. HP55) for oral delivery of insulin. International jour-
[46] Wan F, Møller EH, Yang M, Jørgensen L. Formula- nal of pharmaceutics. 2013; 454(1): 278-84.
tion technologies to overcome unfavorable properties [62] Najafzadeh H, Kooshapur H, Kianidehkordi F. Evalu-
of peptides and proteins for pulmonary delivery. Drug ation of an oral insulin formulation in normal and di-
Discovery Today: Technologies. 2012; 9(2): e141-e6. abetic rats. Indian journal of pharmacology. 2012;
[47] Mohanty RR, Das S. Inhaled Insulin-Current Direc- 44(1): 103.
tion of Insulin Research. Journal of clinical and diag- [63] Wong CY, Martinez J, Dass CR. Oral delivery of
nostic research: JCDR. 2017; 11(4): OE01. insulin for treatment of diabetes: status quo, challeng-
[48] Liu FY, Shao Z, Kildsig DO, Mitra AK. Pulmonary es and opportunities. J Pharm Pharmacol. 2016;
delivery of free and liposomal insulin. Pharm Res. 68(9): 1093-108.
1993; 10(2): 228-32. [64] Yamamoto T. Effects of various protease inhibitors
[49] Malathi S, Nandhakumar P, Pandiyan V, Webster TJ, on the intestinal absorption and degradation of insulin
Balasubramanian S. Novel PLGA-based nanoparticles in rats. Int J Pharm. 1995; 117: 129-37.
for the oral delivery of insulin. Int J Nanomedicine. [65] Tozaki H, EMI Y, Horisaka E, Fujita T, Yamamoto
2015; 10: 2207-18. A, Muranishi S. Degradation of insulin and calcitonin
[50] Patton JS, Bukar JG, Eldon MA. Clinical pharmaco- and their protection by various protease inhibitors in
kinetics and pharmacodynamics of inhaled insulin. rat caecal contents: implications in peptide delivery to
Clinical pharmacokinetics. 2004; 43(12): 781-801. the colon. Journal of pharmacy and pharmacology.
[51] Alabraba V, Farnsworth A, Leigh R, Dodson P, 1997; 49(2): 164-8.
Gough SC, Smyth T. Exubera inhaled insulin in pa- [66] Werle M, Loretz B, Entstrasser D, Föger F. Design
tients with type 1 and type 2 diabetes: the first 12 and evaluation of a chitosan-aprotinin conjugate for
months. Diabetes technology & therapeutics. 2009; the peroral delivery of therapeutic peptides and pro-
11(7): 427-30. teins susceptible to enzymatic degradation. Journal of
[52] Kwok PCL, Chan H-K. Pulmonary delivery of pep- drug targeting. 2007; 15(5): 327-33.
tides and proteins. Peptide and Protein Delivery: [67] Agarwal V, Khan MA. Current status of the oral de-
Elsevier; 2011. p. 23-46. livery of insulin. Pharm Technol. 2001; 10: 76-90.
[53] Richardson PC, Boss AH. Technosphere® insulin [68] Park K, Kwon IC, Park K. Oral protein delivery: Cur-
technology. Diabetes technology & therapeutics. rent status and future prospect. Reactive and Func-
2007; 9(S1): S65-72. tional Polymers. 2011; 71(3): 280-7.
[54] Neumiller JJ, Campbell RK, Wood LD. A review of [69] Egelund R, Rodenburg KW, Andreasen PA, Rasmus-
inhaled technosphere insulin. Annals of Pharma- sen MS, Guldberg RE, Petersen TE. An ester bond
cotherapy. 2010; 44(7-8): 1231-9. linking a fragment of a serine proteinase to its serpin
[55] Klonoff, David C. "Afrezza inhaled insulin: the fast- inhibitor. Biochemistry. 1998; 37(18): 6375-9.
est-acting FDA-approved insulin on the market has [70] Nellans HN. (B) Mechanisms of peptide and protein
favorable properties." 2014: 1071-1073. absorption: (1) paracellular intestinal transport: modu-
[56] Peppas NA, Kavimandan NJ. Nanoscale analysis of lation of absorption. Advanced drug delivery reviews.
protein and peptide absorption: insulin absorption us- 1991; 7(3): 339-64.
Nanotechnology in Insulin Delivery for Management of Diabetes Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 15

[71] Fonte P, Araujo F, Silva C, Pereira C, Reis S, Santos International journal of pharmaceutics. 2005; 293(1-
HA, et al. Polymer-based nanoparticles for oral insu- 2): 271-80.
lin delivery: Revisited approaches. Biotechnol Adv. [85] Li X, Qi J, Xie Y, Zhang X, Hu S, Xu Y, et al.
2015; 33(6 Pt 3): 1342-54. Nanoemulsions coated with alginate/chitosan as oral
[72] Ahmad A, Othman I, Zaini A, Chowdhury E. Oral insulin delivery systems: preparation, characteriza-
nano-insulin therapy: Current progress on nanoparti- tion, and hypoglycemic effect in rats. International
cle-based devices for intestinal epithelium-targeted journal of nanomedicine. 2013; 8: 23.
insulin delivery. J Nanomedic Nanotechnol S. 2014; [86] Sharma G, Sharma AR, Nam JS, Doss GP, Lee SS,
4: 1-10. Chakraborty C. Nanoparticle based insulin delivery
[73] Kaklotar D, Agrawal P, Abdulla A, Singh RP, Mehata system: the next generation efficient therapy for Type
AK, Singh S, et al. Transition from passive to active 1 diabetes. J Nanobiotechnology. 2015; 13: 74.
targeting of oral insulin nanomedicines: enhancement [87] Chalasani KB, Russell-Jones G, Yandrapu SK, Diwan
in bioavailability and glycemic control in diabetes. PV, Jain SK. A novel vitamin B12-nanosphere conju-
Nanomedicine (Lond). 2016; 11(11): 1465-86. gate carrier system for peroral delivery of insulin.
[74] Mukhopadhyay P, Mishra R, Rana D, Kundu PP. Journal of controlled release. 2007; 117(3): 421-9.
Strategies for effective oral insulin delivery with [88] Reis CP, Veiga FJ, Ribeiro AJ, Neufeld RJ, Damgé
modified chitosan nanoparticles: A review. Progress C. Nanoparticulate biopolymers deliver insulin orally
in polymer science. 2012; 37(11): 1457-75. eliciting pharmacological response. Journal of phar-
[75] Zhao L, Su C, Zhu B, Jia Y. Development and Opti- maceutical sciences. 2008; 97(12): 5290-305.
mization of Insulin-Chitosan Nanoparticles. Tropical [89] Woitiski CB, Neufeld RJ, Veiga F, Carvalho RA,
Journal of Pharmaceutical Research. 2014; 13(1): 3-8. Figueiredo IV. Pharmacological effect of orally deliv-
[76] Song L, Zhi Z-l, Pickup JC. Nanolayer encapsulation ered insulin facilitated by multilayered stable nano-
of insulin-chitosan complexes improves efficiency of particles. European Journal of Pharmaceutical Sci-
oral insulin delivery. International journal of nano- ences. 2010; 41(3-4): 556-63.
medicine. 2014; 9: 2127. [90] Yang J, Sun H, Song C. Preparation, characterization
[77] Pan Y, Li Y-j, Zhao H-y, Zheng J-m, Xu H, Wei G, et and in vivo evaluation of pH‐sensitive oral insulin‐
al. Bioadhesive polysaccharide in protein delivery loaded poly (lactic‐co‐glycolicacid) nanoparticles. Di-
system: chitosan nanoparticles improve the intestinal abetes, Obesity and Metabolism. 2012; 14(4): 358-64.
absorption of insulin in vivo. International journal of [91] Wu J-Z, Williams GR, Li H-Y, Wang D-X, Li S-D,
pharmaceutics. 2002; 249(1-2): 139-47. Zhu L-M. Insulin-loaded PLGA microspheres for
[78] Lin Y-H, Chen C-T, Liang H-F, Kulkarni AR, Lee P- glucose-responsive release. Drug delivery. 2017;
W, Chen C-H, et al. Novel nanoparticles for oral in- 24(1): 1513-25.
sulin delivery via the paracellular pathway. Nano- [92] Peng Q, Sun X, Gong T, Wu C-Y, Zhang T, Tan J, et
technology. 2007; 18(10): 105102. al. Injectable and biodegradable thermosensitive hy-
[79] Cui F, Qian F, Zhao Z, Yin L, Tang C, Yin C. Prepa- drogels loaded with PHBHHx nanoparticles for the
ration, characterization, and oral delivery of insulin sustained and controlled release of insulin. Acta bio-
loaded carboxylated chitosan grafted poly (methyl materialia. 2013; 9(2): 5063-9.
methacrylate) nanoparticles. Biomacromolecules. [93] Hosseininasab S, Pashaei-Asl R, Khandaghi AA,
2009; 10(5): 1253-8. Nasrabadi HT, Nejati-Koshki K, Akbarzadeh A, et al.
[80] Elsayed A, Al Remawi M, Qinna N, Farouk A, Bad- Synthesis, characterization, and in vitro studies of
wan A. Formulation and characterization of an oily- PLGA-PEG nanoparticles for oral insulin delivery.
based system for oral delivery of insulin. European Chem Biol Drug Des. 2014; 84(3): 307-15.
Journal of Pharmaceutics and Biopharmaceutics. [94] Cui F, Shi K, Zhang L, Tao A, Kawashima Y. Biode-
2009; 73(2): 269-79. gradable nanoparticles loaded with insulin-
[81] Sarmento B, Martins S, Ribeiro A, Veiga F, Neufeld phospholipid complex for oral delivery: preparation,
R, Ferreira D. Development and comparison of dif- in vitro characterization and in vivo evaluation. Jour-
ferent nanoparticulate polyelectrolyte complexes as nal of controlled release. 2006; 114(2): 242-50.
insulin carriers. International Journal of Peptide Re- [95] Tao A-j, Zhang L-q, Shi K, Cun D-m, Cui F-d. Prepa-
search and Therapeutics. 2006; 12(2): 131-8. ration and characterization of insulin loaded PLGA-
[82] Sarmento B, Martins S, Ferreira D, Souto EB. Oral HP55 nanoparticles [J]. Journal of Shenyang Pharma-
insulin delivery by means of solid lipid nanoparticles. ceutical University. 2007; 1: 001.
Int J Nanomedicine. 2007; 2(4): 743-9. [96] Wang X-Q, Zhang Q. pH-sensitive polymeric nano-
[83] Zhang N, Li J, Jiang W, Ren C, Li J, Xin J, et al. Ef- particles to improve oral bioavailability of pep-
fective protection and controlled release of insulin by tide/protein drugs and poorly water-soluble drugs.
cationic β-cyclodextrin polymers from algi- European Journal of Pharmaceutics and Biopharma-
nate/chitosan nanoparticles. International journal of ceutics. 2012; 82(2): 219-29.
pharmaceutics. 2010; 393(1-2): 213-9. [97] Wu ZM, Zhou L, Guo XD, Jiang W, Ling L, Qian Y,
[84] Ma Z, Lim TM, Lim L. Pharmacological activity of et al. HP55-coated capsule containing PLGA/RS na-
peroral chitosan-insulin nanoparticles in diabetic rats. noparticles for oral delivery of insulin. International
journal of pharmaceutics. 2012; 425(1-2): 1-8.
16 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 2 Bahman et al.

[98] Sun S, Liang N, Piao H, Yamamoto H, Kawashima delivery and the influence of mucus on insulin
Y, Cui F. Insulin-SO (sodium oleate) complex-loaded transport. Biomaterials. 2012; 33(5): 1573-82.
PLGA nanoparticles: formulation, characterization [112] Pridgen EM, Alexis F, Kuo TT, Levy-Nissenbaum E,
and in vivo evaluation. Journal of microencapsulation. Karnik R, Blumberg RS, et al. Transepithelial
2010; 27(6): 471-8. transport of Fc-targeted nanoparticles by the neonatal
[99] Jain S, Rathi VV, Jain AK, Das M, Godugu C. Fo- fc receptor for oral delivery. Science translational
late-decorated PLGA nanoparticles as a rationally de- medicine. 2013; 5(213): 213ra167.
signed vehicle for the oral delivery of insulin. Nano- [113] Hirlekar RS. Oral Insulin Delivery: Novel Strategies.
medicine. 2012; 7(9): 1311-37. Asian Journal of Pharmaceutics (AJP): Free full text
[100] Bock N, Dargaville TR, Woodruff MA. Electrospray- articles from Asian J Pharm. 2017 Oct 11; 11(03)
ing of polymers with therapeutic molecules: state of [114] Arbit E, Kidron M. Oral Insulin Delivery in a Physio-
the art. Progress in polymer science. 2012; 37(11): logic Context: Review. J Diabetes Sci Technol. 2017;
1510-51. 11(4): 825-32.
[101] Damgé C, Maincent P, Ubrich N. Oral delivery of [115] Easa N, Alany RG, Carew M, Vangala A. A review
insulin associated to polymeric nanoparticles in dia- of non-invasive insulin delivery systems for diabetes
betic rats. Journal of controlled release. 2007; 117(2): therapy in clinical trials over the past decade. Drug
163-70. discovery today. 2019; 24(2): 440-451.
[102] Damgé C, Vranckx H, Balschmidt P, Couvreur P. [116] Heinemann L, Jacques Y. Oral insulin and buccal
Poly (alkyl cyanoacrylate) nanospheres for oral ad- insulin: a critical reappraisal. Journal of diabetes sci-
ministration of insulin. Journal of pharmaceutical sci- ence and technology. 2009; 3(3): 568-84.
ences. 1997; 86(12): 1403-9. [117] Chen J, Liu R, Liu C, Jin X, Zhang Q, Wang J, et al.
[103] Hou Z, Zhang Z, Xu Z, Zhang H, Tong Z, Leng Y. Progress of Oral Insulin and Related Drug Delivery
The stability of insulin-loaded polybutylcyanoacrylate Systems and their Pharmacokinetics. Current drug
nanoparticles in an oily medium and the hypoglyce- metabolism. 2018; 19(10): 863-70.
mic effect in diabetic rats. Yao xue xue bao= Acta [118] Li J, Wang Y, Han L, Sun X, Yu H, Yu Y. Time-
pharmaceutica Sinica. 2005; 40(1): 57-64. Action Profile of an Oral Enteric Insulin Formulation
[104] Graf A, Rades T, Hook SM. Oral insulin delivery in Healthy Chinese Volunteers. Clinical therapeutics.
using nanoparticles based on microemulsions with 2012; 34(12): 2333-8.
different structure-types: optimisation and in vivo [119] Čilek A, Čelebi N, Tirnaksiz F. Lecithin-based mi-
evaluation. European journal of pharmaceutical sci- croemulsion of a peptide for oral administration:
ences. 2009; 37(1): 53-61. preparation, characterization, and physical stability of
[105] Müller RH, Mäder K, Gohla S. Solid lipid nanoparti- the formulation. Drug Delivery. 2006; 13(1): 19-24.
cles (SLN) for controlled drug delivery-a review of [120] Elsayed A, Al-Remawi M, Farouk A, Badwan A.
the state of the art. European journal of pharmaceutics Insulin-chitosan polyelectrolyte _anocomplexes:
and biopharmaceutics. 2000; 50(1): 161-77. preparation, characterization and stabilization of insu-
[106] Mehnert W, Mäder K. Solid lipid nanoparticles: pro- lin. Sudan Journal of Medical Sciences. 2010; 5(2):
duction, characterization and applications. Advanced 99-109.
drug delivery reviews. 2012; 64: 83-101. [121] Badwan A, Remawi M, Qinna N, Elsayed A, Arafat
[107] Zhang N, Ping Q, Huang G, Xu W, Cheng Y, Han X. T, Melhim M, et al. Enhancement of oral bioavaila-
Lectin-modified solid lipid nanoparticles as carriers bility of insulin in humans. Neuro endocrinology let-
for oral administration of insulin. International jour- ters. 2009; 30(1): 74-8.
nal of pharmaceutics. 2006; 327(1-2): 153-9. [122] Walsh EG, Adamczyk BE, Chalasani KB, Maher S,
[108] Fonte P, Nogueira T, Gehm C, Ferreira D, Sarmento O’Toole EB, Fox JS, et al. Oral delivery of macro-
B. Chitosan-coated solid lipid nanoparticles enhance molecules: rationale underpinning Gastrointestinal
the oral absorption of insulin. Drug delivery and Permeation Enhancement Technology (GIPET®).
translational research. 2011; 1(4): 299-308. Therapeutic delivery. 2011; 2(12): 1595-610.
[109] Yang R, Gao R, Li F, He H, Tang X. The influence of [123] Zijlstra E, Heinemann L, Plum-Morschel L. Oral in-
lipid characteristics on the formation, in vitro release, sulin reloaded: a structured approach. J Diabetes Sci
and in vivo absorption of protein-loaded SLN pre- Technol. 2014; 8(3): 458-65.
pared by the double emulsion process. Drug devel- [124] Yu J, Qian C, Zhang Y, Cui Z, Zhu Y, Shen Q, et al.
opment and industrial pharmacy. 2011; 37(2): 139-48. Hypoxia and H2O2 dual-sensitive vesicles for en-
[110] Xu Y, Zheng Y, Wu L, Zhu X, Zhang Z, Huang Y. hanced glucose-responsive insulin delivery. Nano let-
Novel solid lipid nanoparticle with endosomal escape ters. 2017; 17(2): 733-9.
function for oral delivery of insulin. ACS applied ma- [125] Wang C, Ye Y, Sun W, Yu J, Wang J, Lawrence DS,
terials & interfaces. 2018; 10(11): 9315-24. et al. Red Blood Cells for Glucose‐Responsive Insulin
[111] Jin Y, Song Y, Zhu X, Zhou D, Chen C, Zhang Z, et Delivery. Advanced Materials. 2017; 29(18):
al. Goblet cell-targeting nanoparticles for oral insulin 1606617.

DISCLAIMER: The above article has been published in Epub (ahead of print) on the basis of the materials provided by the
author. The Editorial Department reserves the right to make minor modifications for further improvement of the manuscript.

You might also like