You are on page 1of 17

Review

Oral insulin delivery: existing barriers and current


counter-strategies
Ahmed Gedawya, Jorge Martineza, Hani Al-Salamia,b and Crispin R. Dassa,b
a
School of Pharmacy and Biomedical Science, Curtin University and bCurtin Health Innovation Research Institute, Bentley, WA, Australia

Keywords Abstract
diabetes; formulation; insulin; oral; tablet
Objectives The chronic and progressive nature of diabetes is usually
Correspondence associated with micro- and macrovascular complications where failure of
Crispin R. Dass, School of Pharmacy, Curtin pancreatic b-cell function and a general condition of hyperglycaemia is
University, GPO Box U1987, Perth, WA created. One possible fac- tor is failure of the patient to comply with and
6845, Australia. adhere to the prescribed insulin due to the inconvenient administration route.
E-mail: Crispin.Dass@curtin.edu.au
This review summarizes the ratio- nale for oral insulin administration,
existing barriers and some counter-strategies trialled.
Key findings Oral insulin mimics the physiology of endogenous insulin
Received August 19, 2017
secreted by pancreas. Following the intestinal absorption of oral insulin, it
Accepted October 24, 2017
reaches the liver at high concentration via the portal vein. Oral insulin on the
other hand has the potential to protect pancreatic b-cells from autoimmune
doi: 10.1111/jphp.12852 destruction. Struc- tural modification, targeting a particular tissue/receptor,
and the use of innova- tive pharmaceutical formulations such as nanoparticles
represent strategies introduced to improve oral insulin bioavailability. They
showed promising results in overcoming the hurdles facing oral insulin
delivery, although delivery is far from ideal.
Summary The use of advanced pharmaceutical technologies and further
research in particulate carrier system delivery predominantly nanoparticle
utilization would offer useful tools in delivering insulin via the oral route
which in turn would potentially improve diabetic patient compliance to
insulin and the overall management of diabetes.

insulin was first trialled in 1922 with poor results. [5]


Introduction Even to this day, a successful oral insulin preparation
Insulin is administered to diabetic patients through the has been elusive, but is expected to dramatically improve
parenteral route. Most protein drugs are administered by patient outcomes. This review will address and discuss
injection due to their perceived low bioavailability if the follow- ing topics: insulin-related physiology and
deliv- ered by other routes. Insulin is injected rationale for oral insulin use, basic challenges in oral
subcutaneously, and this is usually associated with pain, delivery of insulin, some strategies trialled and finally
trauma, distress, leading to poor patient compliance [1,2] mitogenic concerns of oral insulin.
which in turn cas- cades into a chain of undesired
effects. The exogenous insu- lin is significantly able to Insulin-related physiology
reduce the rate of morbidity and mortality, although
around 60% of diabetic patients fail to achieve long-term After it is secreted from pancreatic b-cells into the portal
normoglycaemia,[3] presumably because of non- vein, insulin is directly transferred to the liver where it is
compliance issues resulting from needle phobia and subjected to the hepatic first-pass effect where almost
complexity of the insulin treatment regimen. In effect, half of the polypeptide hormone experiences hepatic
poor patient compliance would contribute to long-term degrada- tion.[6] Such an environment creates an insulin
poor glycaemic control and diabetic ketoacidosis and concentra- tion gradient between hepatic portal and
spark a ser- ies of macro- and microvascular systemic
complications. [4] Oral

© 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213 197
Oral delivery of insulin Ahmed Gedawy et al.

circulation, where the liver is exposed to insulin were taken into consideration by researchers, most of the
concentra- tion twofold to fourfold higher than that developed formulations have failed to demonstrate
observed in the peripheral systemic circulation. [6] satisfy- ing bioavailability results. [7,10]
Available insulin preparations fail to mimic the
endoge- nous insulin pathway where the injectable Oral insulin and rationale for its use
insulin analogues are delivered directly to the
peripheral circulation, hence reversing the insulin
concentration gradient in normal physiology, and as a Physiological rationale
result, insulin reaches the liver at much lower
concentrations than those in non-diabetic individu- Orally administered insulin better replicates the normal
als.[7] Diabetic patients treated with injectable insulin physiological insulin pathway and leads to better
preparations are subjected to the side-effects of glucose homeostasis. [11,12] After its absorption from the
hyperinsuli- naemia, weight gain and hypoglycaemic intestinal lumen, insulin is transported via the portal
risks.[7] circulation to the liver (the ultimate target) [13] creating a
high porto-sys- temic gradient. The oral route offers
decreased levels of sys- temic insulin,[14] hence less
A step towards normal physiology hypoglycaemic episodes and weight gain problems
(Figure 1).
The long-term use of the conventional insulin The liver on the other hand is highly sensitive to
preparations, collectively called multiple daily insulin insulin, and within few minutes, glucose production
(MDI), results in poor glycemic control, risks of by the liver is blocked by a portal insulin
hypoglycaemia and needle phobia. [8,9] concentration of 50 lu/ml in healthy objects and by
On the other hand, treating the dawn phenomenon around 100 lu/ml in diabetic patients.[15–17]
(early morning hyperglycaemia especially in pregnant Interestingly, oral insulin plays a significant role in
patients) with conventional insulin preparations is quite protection of b-cells of the pancreas from autoim-
complex and troublesome. [8,9] The aforementioned draw- mune destruction.[18] Another study concluded the
backs raised the desire of diabetologists to get the basal possi- bility of early insulin therapy to improve b-cell
insulin at a more physiological level through delivering function and provided a chance for b-cell rest.[1,19]
pre- determined continuous subcutaneous insulin Insulin adminis- tered orally theoretically should be
infusion (CSII) or insulin pump which can also get able to overcome the slow first phase release kinetics
programmed to deliver postprandial bolus insulin if encountered with subcuta- neous administration.[1,20]
required.[8,9]
In the late 1970s, insulin pumps were introduced and
it was initially thought to be the most physiological way Pharmaceutical rationale
of delivering insulin subcutaneously to achieve near-
normo- glycaemia. It has been suggested that oral insulin offers induction of
CSII offered more flexibility in daily life and that is oral tolerance or immune modulating effect which is
prob- ably why they were recommended during likely to help in prevention of diabetes. [1,21] The theory
pregnancy or for preconception care and for type 1 behind this phenomenon is that oral administration of a
diabetic children and adolescents.[8,9] New model insulin low dose of antigen alters responsiveness of systemic T
infusion pumps are light in weight (less than 0.5 kg), cell to that antigen which seems to inhibit autoimmune
small in size, and contain replaceable insulin syringes or diseases such as type 1 diabetes.[21] This effect has been
cartridges. The insulin is infused from the pump to the experimented upon non-obese diabetic (NOD) mice
subcutaneous tissue through a simple and manually treated with oral insulin, where peripheral T cells do not
inserted catheter.[8] migrate to pancre- atic islets[21] which might explain
prevention of type 1 dia- betes in NOD mice. [21] Despite
its lower bioavailability, oral delivery of peptides
Less invasive insulin delivery showed high level of acceptance by patients with
improved level of compliance [22] avoiding pain,
Despite all the initial excitement, delivery of insulin discomfort, possible infections by injectable forms in
through MDI or CSII still did not mimic the physiology addition to being less expensive. These are all factors
of the endogenously secreted human insulin, where the that favour a switch to oral delivery of therapeutic
insu- lin is reversely distributed between portal and peptides such as insulin.[22]
systemic cir- culation which has driven researchers to
develop less invasive and more physiological routes of
insulin adminis- tration such as transdermal, inhaled, Challenges in oral delivery of insulin
nasal, buccal, ocular, rectal and the highly preferred oral
insulin.[7] The fundamental barrier to the oral delivery of proteins
While stability requirements for formulation of a is the gastrointestinal tract (GIT). The GIT functions as
protein hormone as well as making sure of its
therapeutic efficacy

198 © 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213
Ahmed Gedawy et al. Oral delivery of insulin

Figure 1 Endogenous versus administered insulin circulation pathways. (a) Endogenous insulin pathway under normal physiology; (b) orally
administered insulin pathway; (c) injected insulin pathway in diabetic patients. [Colour figure can be viewed at wileyonlinelibrary.com]

an absorptive and protective organ. Active macro- B Goblet cells: The second most abundant in the epithe-
molecules have to undergo degradation into inactive lium which acts as a secretory reservoir of
components before they face the next absorption barrier, mucus.[26,27]
the tight epithelium. These natural mechanisms thus also C Paneth cells, when exposed to bacteria, and antimicro-
act as a defence shield against environmental bial proteins are secreted to kill the invading
pathogens.[23–25] bacteria[26,27].
The epithelial cell layer comprises different types of D Microfold cells (M-cells) of Peyer’s patch
cells (Figure 2): characterized by less cytoplasmic lysosomes and lack
A Enterocytes: The most abundant, with dense well- of a mucous layer covering their surface. M-cells take
ordered brush border, has absorptive function via up antigens and microorganisms from the lumen of
trans- porting nutrients.[26,27] the intestine and present these to the immune system
resident in the mucosa.[28,29]

(a)
(f) (g)
(d)

(b)
(c)

(e)

Figure 2 Different cell types of the healthy (normal) epithelial layer. a – enterocytes, b – goblet cells, c – Paneth cells, d – microfold cells (M-cells),
e – paracellular transport, f – transcellular transport, g – carrier-mediated transport. [Colour figure can be viewed at wileyonlinelibrary.com]

© 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213 199
Oral delivery of insulin Ahmed Gedawy et al.

E Paracellular route mainly for small hydrophilic Physical barriers


molecules. 1 The mucous layer: Mucus is the first barrier which
F Transcellular route is limited for passive diffusion of func- tions as both diffusional and enzymatic barrier
lipophilic substrates. encoun- tered by polypeptides. [32,33] Mucus is
G Carrier-mediated transcellular absorption. continuously secreted and probably because of its
negative charge, it filters out positively charged drugs
The tight junctions (TJs) between adjacent epithelial
cells maintain the intactness of the intestinal epithelium and proteins by elec- trostatic interaction. [32–34]
struc- ture. These TJs are composed of various 2 Intestinal epithelium: The intestinal epithelium is
transmembrane proteins such as claudins and com- posed of a single layer of columnar epithelial
occludins.[30,31] cells.[26–29] Passive diffusion is limited to lipophilic
drugs less than 700 Da in molecular weight, [35] and
because the molecu- lar weight of insulin is 5800
Barriers to oral insulin Da,[36] this renders transcel- lular passive diffusion of
insulin into cells very difficult.
Existing barriers could be categorized into three main 3 Tight junctions: This dynamic structure is selectively
sub- types, namely physical, biochemical and per- meable to small hydrophilic molecules (nutrients,
formulation-based. (Table 1 summarizes possible ions
barriers for oral insulin deliv- ery and some of the
strategies used).

Table 1 Types of barriers facing oral insulin administration and counter-strategies

Examples of agents used


Barrier Strategies introduced and/or contributing factor Major findings References

Mucus layer Mucoadhesive Chitosan, alginate, Mucoadhesive polymers provide [32,33,83,85]


systems polyacrylic acid an intimate contact with the
mucosa at the drug uptake site
PLGA, thiomers The strategy increases the residence
time of the delivery system at the
absorption site
Mucus Hydrophilic polymer coating Efficient mucus permeation [87–89]
penetration systems such as (PEG), Virus-mimicking requires:highly densely charged
strategy (highly charged surface) surface, Hydrophilic shell and
slightly negative surface charge
Intestinal epithelial Carrier-mediated Transferrin, immunoglobulins Due to their large molecular weight, [45,53,56,58]
cells and the transcellular biotin, lectins, folate, conjugation of proteins to
transcellular absorption vitamin B12 receptor-recognisable ligands
route will have the potential to improve
cellular internalisation
Cell penetrating Penetratin, oligoarginine, Perturbing cell membrane, endocytosis [59–61]
peptides (CPPs) transportan, TAT or even channel formation could be
a possible mechanism of their action
Absorption enhancers EDTA, surfactants, Perturb the cell membrane [44,46,69,71]
fatty acids, ZOT
Tight junction Absorption enhancers Selectively open tight junctions
and paracellular Particulate carrier Lipid suspension, liposomes, They circumvent the harsh gastric [28,29,90,91,99–104]
route systems submicroemulsion, conditions without affecting the
nanospheres, bioactivity of macromolecules
SLN, HDV-I, SNEDDS like insulin
M cells of Peyer’s patches play a
unique role in the uptake of small
particles
Luminal pH Colon-targeting and CODES, capsulin, Low level of luminal and brush [56,73–79]
enteric coating ORMD-0801 border proteases
Enzymatic Protease inhibitors Na-glycocholate, soya bean trypsin Concerns about long-term use [46,67,68]
degradation inhibitor, aprotinin, bacitracin, and protein malabsorption
camostat mesilate,
chicken and duck ovomucoids
PLGA, polylactic co-glycolic acid; PEG, polyethylene glycol; EDTA, ethylene diamine tetraacetic acid; ZOT zonula occludens toxin; SLN, solid lipid
nanoparticle; HDV-I, hepatic direct vesicle insulin; SNEDDS, self-nanoemulsifying drug delivery system.

200 © 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213
Ahmed Gedawy et al. Oral delivery of insulin

and certain drugs).[30,31] Solutes of molecular radius The suggested strategies trialled for oral insulin
more than 15 Å are usually excluded from traversing delivery can be classified into:
this barrier.[31] 1 Chemical modification
The covalent attachment of polyethylene glycol (PEG)
to therapeutic peptides has been utilized previously; the
Biochemical barriers tech- nology is called PEGylation. [48] PEGylation has
1 Luminal pH: pH varies from highly acidic (1.2–3.0) been used to decrease the rate of clearance and improve
in the stomach to slightly basic (6.5–8.0) in the the pharma- cological and biological properties of
intestine.[37] Such pH variation can cause pH- peptides and elimi- nates the immunogenicity,
induced oxidation and deamination of protein allergenicity and antigenicity of insulin if compared with
drugs.[38] unmodified subcutaneous insu- lin.[49] (PEG-INTRON®)
2 Enzymatic degradation: Proteolysis starts at the or PEG-modified a-interferon is an example of this
stomach through the action of pepsin and continues technology which has been approved for hepatitis C
throughout the intestine due to chymotrypsin, elastase treatment.[50]
and car- boxypeptidases.[39] Presystemic degradation The NOBEX Corporation worked on the same
of proteins could also be attributed to cytosolic and principal to develop hexyl insulin mono-conjugate-2
membrane- bound enzymes of the enterocytes. [39] (HIM2). A short- chain PEG is linked to a lipophilic
alkyl group to produce an oligomer. The amphiphilic
Insulin is predominantly degraded by trypsin, a- oligomer was used to modify recombinant human
chymo- trypsin and carboxypeptidases in the mucous insulin through attachment to lysine at position 29 of
layer as well as intestinal lumen, with research also the B-chain[51] (Figure 3). HIM2 showed increased
showing the exis- tence of a specific insulin-degrading solubility, absorption, good stability against enzy- matic
enzyme (IDE) on the brush-border membrane.[40,41] degradation, although oral bioavailability was still ~5%
Peptide drugs that do cross the intestinal epithelial and clinical studies showed efficacy in both type I and
bar- rier then undergo further degradation in the liver type II diabetic patients.[52,53] The research into
(hepatic first-pass metabolism).[1,42] PEGylated insulin technology has also been pursued by
It has initially been thought that the inhibition of the Biocon, a large Indian pharmaceutical company. Biocon
IDE could be of value in preserving oral insulin took over NOBEX in 2005 and acquired the intellectual
formula- tions from enzymatic degradation, thereby property of HIM2 where they developed the oral insulin
improving its oral absorption and efficacy. A novel candidate (IN-105) as a second- generation tablet.[54–56]
potent IDE inhibitor (6bK) has been revealed in a recent IN-105 had an improved stability profile in GIT and
study which unfortu- nately did not improve oral insulin enhanced absorption and in comparison with normal
delivery. [43] IDE inhibi- tion has been associated with insulin, lower immunogenicity, lower mito- genicity
increased amylin levels, which in turn slows the gastric and the same pharmacological action.[55]
emptying rate and improves glu- cose tolerance.[43] Thus, Another example of structural modification has been
although the aim of improving oral insulin absorption pioneered by Emisphere’s EligenTM. The technology
was not attained, IDE inhibitors can still be of benefit in com- prises a reversible non-covalent interaction of
the treatment of T2DM patients.[43] insulin with low molecular weight carriers (200–400
Da).[45] These car- rier molecules are organic/lipophilic
Formulation barrier in nature to improve the lipophilicity of insulin and
facilitates passive transcellu- lar diffusion without
The fabrication method could be the last barrier in affecting cell membrane integrity or TJs.[45] Despite the
formu- lation of peptide drugs. Being a sensitive relative fast absorption rate obtained from the
polypeptide hor- mone and any conformational changes pharmacokinetic data, Emisphere did not show a
to insulin structure would affect its biological satisfactory bioavailability even in the presence of the
activity.[44] Better ways for formulation of oral peptide large amount of carriers needed per dose.[45,55,57]
drugs have been tack- led by many scientists through 2 Targeting receptor/tissue
various approaches, with two foci being the overcoming
of intestinal hurdles and the ensuing low a Receptor-mediated endocytosis: It is thought that
bioavailability.[45,46] Co-administration of enzyme exploiting endogenous cellular transport systems
inhibitors, addition of absorption enhancers or even could increase uptake of some drugs.[45,53] Cell mem-
slight modifications of the chemical structure of the brane transporters have been utilized previously to
protein are all among these techniques. [45,46] These trans- port relatively small therapeutics. Receptor-
emerg- ing techniques (below) are basically aiming to mediated endocytosis, on the other hand, offered better
improve pharmacokinetic and pharmacodynamic results in transport of macromolecules and proteins.[53]
characteristics of the hormone.45–47 Lectins, transferrin, immunoglobulins, folate,
cyanocobalamin (vitamin B12, vit B12), epidermal
growth factor and

© 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213 201
Oral delivery of insulin Ahmed Gedawy et al.

Figure 3 Hexyl insulin mono-conjugate-2 (HIM2). A short-chain polyethylene glycol (PEG) is attached to lysine at position 29 of the insulin B-
chain. [Colour figure can be viewed at wileyonlinelibrary.com]

intrinsic protein complex are among receptor- 3 Formulation/pharmaceutical technologiesThe use of


recog- nizable ligands that have been covalently these formulation technologies/strategies does not
attached to a peptide to improve both specificity include physicochemical modification of the
and intracellular delivery.[45,53] This principle has peptide to withstand gastric conditions. In fact,
been adopted by Access Pharmaceuticals in shielding insulin by encapsulation in an enteric-
development of CobOralTM, a peptide-loaded coated dosage form, co- administration with
dextran nanoparticle coated with cobalamin. protease inhibitor or addition of permeation
Likewise, Apollo Life has developed a car- enhancers have been deployed with varying degrees
bohydrate-based nanoparticle coated with Vit
TM
of success. Detergents, fatty acids or bile acid salts
B12 called Oradell [56]. Vit B12-coated dextran are among the absorption enhancers used to facili-
nanoparti- cles loaded with insulin showed tate the internalization of the hormone and
significant prolonged hypoglycaemic effect in a overcoming the intestinal conditions to some
STZ diabetic rat model.[58] extent.[44–46,56] Exam- ples of formulation-related
b Cell-penetrating peptides (CPPs): CPPs is a technologies are as follows:
relatively recent strategy in its early stage which
has the poten- tial for effective delivery of a Gut-associated lymphoid tissues: Gut-associated lym-
liposomes, nanoparticles, small molecules, as well phoid tissues are best represented by Microfold cells
as peptide drugs. [59] The technique is based upon (M-cells) of Peyer’s patch. M-cells are responsible for
using a class of short pep- tides such as penetratin, antigen sampling and microparticulate uptake smaller
oligoarginine, transportan and HIV-1 transactivator than 10 lm.[63,64] An uptake that has been
of transcription (TAT). [60] No clear mechanism of used before for mucosal vaccine delivery through the
translocation and internaliza- tion of their cargoes per- oral route.[63] Nanoparticles of chitosan showed a
into the cytoplasm has emerged, although direct sig- nificant protection of encapsulated insulin against
perturbation of the cell membrane, endocytosis or enzymatic degradation, and such nanoparticles were
channel formation is possible mecha- nisms that able to cross the epithelium through Peyer’s
have been hypothesized. [61] CPPs showed no patches.[65,66]
toxicity or side-effects with in-vivo use and the b Protease inhibitors: As insulin is degraded by trypsin,
transport of insulin/TAT conjugate across Caco-2 a-chymotrypsin and elastase, inhibition of the activity
cells showed significant improvement.[53,62]

202 © 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213
Ahmed Gedawy et al. Oral delivery of insulin

of these enzymes would decrease presystemic bioavailability. Polyacrylic-coated gelatin capsules


insulin degradation and improve its absorption. [46] loaded with insulin have been studied in rats and
Long- term use of these inhibitors is still an issue showed significant drop in the blood glucose level
which could result in protein malabsorption due to compared to intraperitoneal injection. [72] An
dis- turbed protein digestion. Protease inhibitors interest- ing colon-specific drug delivery system,
could also be associated with systemic toxicity. [44] CODESTM, is based on a core tablet coated with
To this end, the enzyme inhibitory effect of Na- different polymeric layers. CODES showed
glycocholate, aprotinin, bacitracin, soya bean sustained release of insulin in the colon of dogs.
trypsin inhibitor and camostat mesilate has been The formula incorporated the hor- mone with
compared. Co-adminis- tration of these agents with meglumine (a pH adjuster), citric acid (insulin
insulin directly into iso- lated intestines of normal solubilizer), Na-glycocholate (a permeation
rats resulted in improvement of the bioavailability enhancer) along with polyethylene oxide. [73,74]Cap-
of insulin. The effect was more predominant in the sulinTM, a product of diabetology, is an enteric-
large intestine than the small intestine where Na- coated capsule loaded with dry powder mixture of
glycocholate, baci- tracin and camostat mesilate insulin, permeation enhancer and solubilizer. The
showed better results.[67] Chicken ovomucoid ingredients of the capsule are generally safe
(Ckovm) and duck ovomucoid (Dkovm) also pharmacopoeial excipients. [75–77] Sixteen T2DM
showed protective effect against trypsin and a- patients were sub- jected to 11 days apart, two
chymotrypsin degradation of insulin. This study isoglycaemic glucose clamp studies in which they
concluded that Ckovm inhibited trypsin- mediated compared the pharma- cokinetic and
degradation at 1 : 1 ratio, while Dkovm showed pharmacodynamic outcomes of both (150U or
100% protection against trypsin and a-chy- 300U) of orally ingested Capsulin and 12U of
motrypsin for 60 min at 1 : 2 ratio of enzyme subcutaneously injected regular human insulin
inhibitor.[68] (Actrapid),[77] and Capsulin showed good gastric
c Absorption enhancers/permeation enhancers (PEs): sta- bility where the dissolution starts in jejunum,
PEs are a group of agents that promote absorption reason- able safety profile with well-tolerated
of therapeutics through perturbing the cell statistically significant hypoglycaemia for over 6
membrane to improve transcellular transport or by h.[75–77]Another pharmaceutical company, Oramed,
selective action on TJs to enhance paracellular has also devel- oped an enteric-coated capsule
permeability. Decreas- ing mucous fluidity, change (ORMD-0801) con- taining 8 mg insulin which has
in membrane fluidity and leakage of proteins by been marketed for type I and type II diabetic
opening TJs are possible mech- anisms suggested patients. [78,79] The pharma- cokinetic and
for their action.[46,69] However, the long-term use of pharmacodynamic characteristics of ORMD-0801
PE is a controversial issue, as some researchers are capsules were evaluated through phase IIa when 2
concerned about toxic effects upon prolonged capsules of the product were given to type 1
use.[46,69] However, others argue that their use is by diabetic patients. [76] The study concluded the safety
far the most advanced technology in oral peptide and biological activity of the product when given
delivery. They reckon clinical studies showed good before meals, and it is eliminated in around 5 h. [76]
safety profile and achieved consistency of Phase IIb study has been conducted in type 2
bioavailability for peptides less than 10k Da. [70] patients to also determine how safe, tolerable and
Bile salts, ethylene diamine tetraacetic acid, effective ORMD-0801 is during 6-week period
surfactants, fatty acids and zonula occludens toxin when com- pared to placebo.[76] ORMD-0801
(ZOT) are examples of permeation enhancers showed remarkable reduction in blood glucose
commonly used to improve oral peptide level, increased plasma insulin level as well as
bioavailability.[44,46,69] In-vivo results showed that reduction in C-peptide.[78,79]A promising approach
ZOT increased insulin oral absorption 10-fold from in oral peptide delivery is the introduction of
rabbit ileum and jejunum and no effect was superporous hydrogel (SPH) and superporous
noticed in colon.[71] hydrogel composite (SPHC) in targeting proteins
d Site-specific deliveryColon targeting has been inter- to a specific site of the intestine. [80] The new core
estingly used to deliver therapeutic peptides and and shuttle delivery system were developed to
non-peptides. Low level of luminal and brush- mechanically localize the peptide drugs in the
border proteases compared to duodenum and intesti- nal lumen.[80] The drug is incorporated in
jejunum encouraged scientists to the use of colon the core which is either attached to or embedded in
targeting to circumvent harsh gastric conditions. [56] the enteric-coated shuttle or conveyor of SPH and
Insulin has been encapsulated by pH-sensitive SPHC.[80] The novel delivery system tried to
materials to con- trol its release and that showed achieve an in-vitro double-phase time-controlled
improved oral release model and showed partial inactivation of
trypsin due

© 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213 203
Oral delivery of insulin Ahmed Gedawy et al.

to calcium binding and enzyme entrapment within for mucus penetration systems. [87] The specific
the polymeric system. [80] Interpenetrating design, PEG molecular weight requirements,
polymeric networks of superporous hydrogels limited cellular uptake and induction of anti-PEG
(SPH-IPNs) using poly (acrylic acid-co- antibodies might be considered potential limitations
acrylamide) and O-carboxy- methyl chitosan have of PEGyla- tion technology. [88] N-(2-
been trialled to estimate insulin transport across rat hydroxypropyl) methacry- lamide (HPMA) is
intestine and colon.[81] No con- formational another hydrophilic mucus inert polymer, and
changes to insulin or alterations to its oral HPMA has been recently used to coat insulin-
bioactivity in terms of hypoglycaemic action have loaded N-trimethyl chitosan (Ins-TMC)
been recorded using (SPH-IPNs) following nanocarriers. The novel approach utilized the
adminis- tration to healthy animals. [81]When it is mucus penetration characteristics of HPMA to
compared to the subcutaneous route, insulin-loaded deliver the mucoadhesive (Ins-TMC) nanocarriers.
SPH-IPNs delivery system achieved around 4% Upon oral administration to diabetic rats,
pharmacological availability and the system partly remarkable hypogly- caemia has been noticed with
inactivated trypsin and a-chymotrypsin.[81] This a potential success of oral insulin delivery.[89]
system improved para- cellular insulin transport f Particulate carrier systemFormulation of drugs with
through excised rat intestine and across CaCo-2 colloidal particulate carriers has been widely used
cell monolayers by 4.2- and 4.9- fold, respectively, to improve peptide delivery. Submicroemulsion,
via transient opening of epithelial TJs, and it can lipid suspension, liposomes, polymeric micro- and
be retained in rat intestine for longer compared to nanoparticles and polymeric micelles are good
the powdered SPH-IPN.[82] exam- ples of this.[90,91] These approaches seem to
e Bioadhesive (mucoadhesive) systems and mucus circum- vent the existing barriers and harsh gastric
pen- etrationThe mucoadhesive properties of conditions for oral peptide delivery demonstrating
some poly- mers have been exploited to prolong that good release profiles and the bioactivity of
the residence time of the drug at its absorption hormones such as insulin could be well maintained
site by increasing the contact with mucosa which too.[90,91] Encap- sulation of insulin in the form of
in fact increases the concentration gradient of the nanospheres facili- tates its absorption across the
drug.[83] Some of these polymers in addition to intestinal epithelium. The nanospheres are taken up
their mucoadhesive charac- teristics have a dual in tissues such as liver where polymer degradation
role as protease inhibitors or per- meation occurs.[52] Insulin nano- spheres could then function
enhancers.[83,84] A new generation of as minor reservoirs for insulin inside the targeted
mucoadhesives was formulated via the organ (the liver). [52,64,92] Chitosan microspheres
introduction of thiomers or thiolated polymers. loaded with insulin have been studied too. Relative
Thiomers have dramatically improved the increase in insulin pharmacologi- cal bioavailability
mucoadhesive properties of anionic polymers has been observed when optimal particle size of the
such as polyacrylic acid and algi- nate or cationic microspheres is selected.[93]
polymers such as chitosan.[85] This improvement
is attributed to the formation of a stronger A novel insulin-loaded chitosan phthalate microsphere
disulphide bond which can improve the formulation was found to sustain the plasma glucose of
mucoadhesive properties 140-fold compared to rats for at least 16 h at the prediabetic level with
unmodified polymers.[85] Chitosan-4- improved oral bioavailability. [94] Another novel solid-in-
thiobutylami- dine insulin-loaded tablets showed oil-in-water (S/O/W) emulsion has been formulated with
controlled release in non-diabetic rats for over 8 reasonable efficacy in oral delivery of peptides and
h.[86]Mucus penetra- tion is another technology proteins. This technique is used to encapsulate insulin
introduced to overcome the dynamic upstream and showed a pH-responsive release pattern simulating
mucus barrier resulted from its rapid turnover GIT condi- tions.[95] In an interesting technique based on
kinetics.[87–89]Efficient mucus penetra- tion of an a low shear reverse micellar approach, a surfactant
insulin carrier requires highly densely charged mixture was pre- pared by mixing
surface (virus-mimicking strategy), coating of didoceyldimethylammonium bromide (a surfactant) and
insulin carrier with hydrophilic mucus inert propylene glycol (a co-surfactant). Tri- acetin (the oily
polymer and neutral to slightly negative charged phase) was then added to the surfactant mixture at the
surface (to minimize electrostatic interaction with optimized ratio of 1 : 3, respectively. The aqueous
mucus).[87]PEGylation is one of the approaches insulin solution was eventually added to the previ- ous
that used PEG coat as hydrophilic mucus inert mixture while stirring to make a final concentration of
polymer to facilitate mucus penetration of 20% v/v aqueous phase in the mixture. [96] The
insulin-loaded carri- ers.[88] Virus-mimicking transparent microemulsion produced was able to entrap
strategy has been combined with PEGylation insulin to
technique in a study for oral insulin delivery as a
model of utilizing two different concepts

204 © 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213
Ahmed Gedawy et al. Oral delivery of insulin

considerable extent (~85%) without affecting its serum insulin level showed pronounced increase in in-
conforma- tional stability. In-vivo results showed 10- vivo studies when compared to oral insulin solution. [107]
fold improve- ment in bioavailability compared to SNEDDS loaded with insulin-lecithin complex
plain insulin solution upon oral administration to facilitated the transport of insulin phospholipid complex
healthy rats.[96] across Madin–Darby canine kidney (MDCK) cell
Limited success since 1976 has been achieved using monolayer, with no cytotoxicity. [108] Oral administration
liposomes for oral delivery of peptides. [97,98] The unsatis- to diabetic Wistar rats resulted in remarkable
factory bioavailability with liposomes could be hypoglycaemia with relative bioavailability up to
attributed to their instability or limited absorption in GIT 7.15%.[108] In a recent study, a novel SNEDDS was
condi- tions, although the natural chemicals developed to facilitate the mucus permeation of oral
(phospholipids) used for formulation are a great insulin,[109] based upon hydrophobic ion pair of insulin
advantage.[97,98] How- ever, a liposomal insulin with dimyristoyl phosphatidylglycerol (DMPG) to form
formulation known as hepatic direct vesicle insulin insulin/DMPG complexes. [104,109]
(HDV-I) has been successfully deliv- ered orally.[55,99,100] The technique relied on the fact that insulin acquires
The novel vesicles are composed of (˂150 nm diameter) net positive charge at low pH which facilitates its
liposomes which contain insulin attached to a specific conjugation with anionic amphiphilic partners
proprietary molecule known as hep- atocyte-targeting (phospholipids, surfac- tants and fatty acids) through
molecule in an attempt to replicate nor- mal insulin ionic complexation. [104,109] In addition to the good
physiology. [55,99,100] HDV-I is formulated as an oral permeation characteristic, insulin/ DMPG complex-
gel capsule, stable at low pH of blood and resists gastric loaded SNEDDS provided protection from GIT
degradation with high biopotency. enzymes and prevented initial burst release of insulin
The solid lipid nanoparticle (SLN) is another example which demonstrates a potential for oral insulin
of lipid-based nanoparticulate carrier that has been delivery. [104,109]
developed as an alternative to polymeric nanoparticle
systems. [101,102] SLNs show good biocompatibility, Nanoparticle architecture and polymer
biodegradation, sus- tained release of the incorporated
molecules and amenabil- ity to large-scale choice in oral insulin delivery
production.[101,102] Insulin-loaded PEG-stearate-coated
Nanotechnology using polymers has been one of the
lipid nanoparticles offer good protection of the
most interesting approaches in formulation of peptide
encapsulated insulin when in contact with gastrointesti- nal
therapeu- tics, and to this end, insulin formulated using
fluids compared to non-coated lipid nanoparticles.[103]
nanoparticle technology showed increased cellular
Self-nanoemulsifying drug delivery systems
uptake and transport across Caco-2 cells.[110] It has been
(SNEDDS) are recent techniques which combine the
the focus of many researchers to optimize nanoparticle
advantages of nanotechnology and lipid-based drug
carriers to protect macromolecules from gastric enzymes
delivery. [104] The strategy was introduced to improve the
as well as improve intestinal permeation. [56] Different
formulation of not only the lipophilic drugs but poorly
studies have been car- ried out using polymer-based
water-soluble thera- peutics as well.[104] SNEDDS were
nanoparticles for oral insulin delivery using different
initially introduced to overcome the drug solubilization
polymers and their derivatives, and the results have been
and bioavailability issues of biomolecules due to their
quite promising [111] (Table 2).
ability to enhance droplet sur- face area, protect from
Generally, an ideal polymeric carrier should be
gastric enzymes, improve perme- ation, alter gastric
biodegrad- able, biocompatible and able to prolong
retention time and sometimes drug mucoadhesive
intestinal residence time after resisting gastric pH
properties. [104] Current applications of SNEDDS include
gradient and enzymes.[112]
improving drug stability, controlling the drug release,
Based on nature, polymers utilized in fabrication of
increasing drug loading capacity (<25 mg to
insu- lin nanoparticles are either of natural or synthetic
>2 g) and specific tissue targeting which can be of
ori- gin.[111,112] Gelatin and casein, examples of natural
potential in lymphoma, leukaemia and autoimmune
protein polymers, have been used to prepare insulin-
diseases.[104,105] In a formulation development study,
loaded nanoparticles for per-oral delivery with
SNEDDS was pro- duced using a solid dispersion
promising results and good safety profile. [112] Natural
technique in a trial to for- mulate a non-invasive
polysaccharide poly- mers have also been used
delivery carrier for therapeutic proteins and it showed
extensively in nanoparticle design of insulin carriers due
promising results.[106] In an attempt to improve oral
to their non-toxic characteristics. Examples of these
insulin bioavailability, researchers com- bined the
polysaccharides are chitosan, alginate, dextran, starch
technology of a multifunctional polymeric system using
and pectin.[112]
thiolated chitosan and SNEDDS technology to pro-
The use of synthetic polymers in formulation of oral
duce (80–160 nm) spheres with remarkable insulin insulin has been advantageous in sustaining the release
entrap- ment efficiency. [107] In-vitro release profile of insulin over a period of days to several weeks
from insulin/ thiolated chitosan SNEDDS compared to natural polymers. [111] Examples of these
significantly increased and synthetic polymers

© 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213 205
Oral delivery of insulin Ahmed Gedawy et al.

Table 2 Examples of polymeric nanocarriers used for oral insulin formulation and their efficacy

Nanoparticle Dose
size (nm) Animal model (IU/kg) Comment References

Poly lactic-co-glycolic 200 STZ diabetic 20 Up to 90% insulin entrapment efficiency [129]
acid (PLGA) rats 7.7% relative bioavailability compared to
subcutaneous route
Prolonged effect (12 h)
57.4% reduction in plasma glucose level within first 8 h
Poly lactic acid 56 Diabetic mice 50 Insulin loading capacity of 0.08% weight [130]
(PLA-F127-PLA) Hypoglycaemia over 23 h
Highest blood glucose reduction
(from 18.5 to 4.5 mmol/l) after 5 h
Poly (Ԑ-Caprolactone)/ 700 Diabetic rats 50 97.5% insulin encapsulated with 70% in [131]
Eudragit RS vitro release over 24 h
Peak hypoglycaemia at 8 h (—52% for
nanoparticles loaded with Aspart insulin)
Dextran + Vit B12 150–300 STZ diabetic 20 Prolonged antidiabetic effect (54 h) [58]
rats Blood glucose reduction of 70–75%
29.4% relative bioavailability compared to
subcutaneous injection
Cetyl palmitate-based 350 Diabetic rats 50 >43% association efficiency [132]
solid lipid Considerable hypoglycaemia over 24 h
nanoparticle (SLN) Relative bioavailability of 5.1% compared to
subcutaneous route
Chitosan 250–400 Diabetic 21 Prolonged hypoglycaemia for >15 h [65]
Wistar rats Relative bioavailability for up to 14.9% compared
to subcutaneous route
Chitosan/Alginate 750 STZ diabetic 50 and Association efficiency > 70% [133]
rats 100 >40% reduction in serum glucose level for >18 h
Relative bioavailability of: 6.8% for 50 IU/kg
and 3.4% for 100 IU/kg compared to
subcutaneous injection
Chitosan/poly ɣ- 218 Diabetic rats 30 71.8% loading efficiency [134]
glutamic acid Significant hypoglycaemia for not less than 10 h
Relative bioavailability for up to 15.1% compared
to subcutaneous route

are polylactic acid, polylactic co-glycolic acid and poly The surface of nanoparticles exhibits high reactivity
(Ԑ-caprolactone), and they all have hydrophobic nat- towards proteins, biomolecules and biological fluids in
ure.[111,113] The internalization and uptake process of the cell interior which cause these subcellular structures
nanoparticles via the transcellular pathway is dependent to asso- ciate with the nanoparticle surface in a
on different factors such as surface charge, phenomenon called bimolecular corona formation.[120]
mucoadhesive properties and particle size. [114,115] Due to The design, chemical composition, size, shape,
their biocompat- ibility and low toxicity, gold solubility, surface functionality and aggregation state of
nanoparticles have been used to deliver insulin orally nanoparticles are all important factors in controlling and
and intranasally. Hypoglycaemia has been recorded determining not only the cellular and subcel- lular
when given to diabetic Wistar rats.[7,116] transport of nanoparticles but their biokinetics and
Cytotoxicity test as well as immunological response their biodistribution as well.[118,120,121]
could be a requirement to assess the safety of Postinternalization of nanoparticles inside the cells,
nanoparticles. Biodegraded nanoparticles if their surface charge can potentially determine the
accumulating inside the cells can cause intercellular targeted sub- cellular organelle (lysosomes, cytoplasm,
changes in terms of organelle integ- rity which could mitochondria or even the nucleus). [120,122] Possible
lead to severe toxicity.[117] It has been sug- gested to cellular toxicity of NPs is best explained through the
consider the nanomaterial waste as unsafe due to their reactive oxygen species and oxidative stress
behaviour at the cellular and subcellular levels. [118] model[120,123] which creates a vicious envi- ronment such
Inadequate data about the potential toxicity of as perturbation of mitochondrial activity, induction of
nanomateri- als to cellular and subcellular organelles are pro-inflammatory effects and nuclear uptake. [118,121,123]
the main reason for the limited use of many All these effects could eventually lead to
nanoparticles.[119]

206 © 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213
Ahmed Gedawy et al. Oral delivery of insulin

cell injury and death [118,121,123] (Figure 4). This perturbed in the enteric-coated capsule offered mucoadhesion to
activity caused by NPs has been labelled by some lar- ger surface area of the intestinal mucosa and resulted
scientists as latent toxicity due to modulation of in a significant hypoglycaemia to male Wistar rats
otherwise normal cellular activity. [119] following oral administration.[125]
In a proof-of-concept study, insulin-loaded dodecy-
Recent developments in oral insulin delivery lamine-graft-ɣ-polyglutamic acid micelles were
developed and cross-linked with trimethyl chitosan
A novel intestinal insulin device has been developed (TMC) in the form of nanoparticle complex. The novel
from a mixture of mucoadhesive polymers in the form of self-assembled polyelec- trolyte complex nanoparticles
13-mm disc. The disc has been coated three times with are further modified to improve their affinity to the
ethyl cellu- lose leaving one surface uncoated to ensure epithelium. This has been achieved by attachment of
unidirectional release of its insulin load. The goblet cell targeting peptide (CSKSSDYQC) to TMC
mucoadhesive disc along with dimethyl palmitoyl coat. Oral administration of the developed targeted
ammonio propanesulfonate (a permeation enhancer) has nanoparticles has a relative bioavailability of 7.05% with
been placed in a capsule (enteric coated with Eudragit prolonged hypoglycaemia in diabetic rats.[126] In a novel
L100) for intestinal delivery. After it has been liberated attempt to modify the hydrophobic nature of SLNs and
from the capsule shell, the insulin-loaded device improve insulin encapsulation efficiency, metho- cel, a
completely released its protein content within 4 h with hydrophilic polymer has been incorporated into the
promising results in the animal model.[124] internal phase of w/o/w double emulsion based SLN.
The same technique has been exploited in the Methocel-lipid hybrid nanocarrier (MLN) has been
develop- ment of 500 lm-sized intestinal insulin pre-
patch. The novel micropatch (either coated or pared by emulsification solvent evaporation technique.
uncoated) has been encapsu- lated the same way with In addition to the improved encapsulation efficiency,
dimethyl palmitoyl ammonio propanesulfonate in MLN also offered extra stability to entrapped insulin
addition to citric acid (a non-specific protease with the potential for a promising nanocarrier for oral
inhibitor). The small size of the patches delivered peptide delivery.[127]

Nanoparticles (NPs)

Generation of reactive Activation of macrophages


oxygen species (ROS) (inflammatory cells)
Oxidative
stress

Perturbation of
Phagocytic function
Activation of pro-
inflammatory
signaling cascade

↓NPs clearance and ↑


their toxicity

↑ Inflammation
Perturbation of Nuclear DNA
mitochondrial function oxidative damage

Impairment of electron transport Cell cycle arrest, blocking


chain, energy failure, permeability of replication/division
transition pore opening and
cytotoxicity

Apoptosis Mutagenesis

Figure 4 Currently known cytotoxic effects of nanoparticles. [Colour figure can be viewed at wileyonlinelibrary.com]

© 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213 207
Oral delivery of insulin Ahmed Gedawy et al.

In a recent study, polyelectrolyte complexation of two potential to alleviate diabetes-associated oxidative stress
natural polymers chitosan and carboxymethylated iota- and improve pancreatic b-cell functions. [130]
carrageenan has been employed in the production of In a recent study based on core-shell nanoparticle
insulin-loaded nanoparticles. The carboxymethylation of technology, insulin has been encapsulated in polyurethane-
iota-carrageenan as well as nanoparticle formulation has alginate core with an outermost shell of chitosan. Insulin-
been optimized through the computational model, loaded polyurethane-alginate/chitosan nanoparticles
response surface methodology based on Box–Behnken controlled the release of the entrapped hormone and the
design. Insulin has pH-responsive release from mucoadhe- sive nature played an important role in the
chitosan/carboxymethylated iota-carrageenan hypoglycaemic effect noticed on fasted Swiss albino
nanoparticles with good protection against gastric mice. The histopatho- logical studies suggest the safe use
conditions. [128] of the developed nanopar- ticles to deliver insulin
Due to the growing interest in developing starch- orally.[131]
based nanocarriers, short-chain glucan, SCG (a In a recent study, hydrophobic ion pairing technology
debranched starch), has been employed in manufacture has been employed to complex insulin with sodium
of insulin–SCG nanoparticles. The addition of cross- deoxycholate. Insulin-sodium deoxycholate complex
linker, proanthocyani- dins (PAC) (isolated from red improved the liposolubility of insulin and enhanced
peanut skins) improved the insulin encapsulation encapsulation efficiency of insulin complex in poly(lac-
efficiency. Insulin SCG/PAC nanoparticles exhibited tide-co-glycolide) (PLGA). Insulin-loaded PLGA
gastric stability and significant hypoglycaemia for 8 h nanopar- ticles were prepared by emulsion solvent
following oral administration in dia- betic rat model.[129] diffusion method. PLGA nanoparticles are then spray-
In a proof-of-concept study, insulin-loaded selenium dried with hydroxypropyl methyl cellulose phthalate (a
nanoparticles were formulated by ionic cross-linking pH-respon- sive polymer) for intestinal delivery. The
reduc- tion technique. Insulin-selenium nanoparticles new multifunc- tional composite microcapsule
were pro- duced in situ. Addition of sodium selenite and represents a promising carrier for oral peptide
glutathione onto insulin/chitosan complex caused delivery. [132]
reduction in sele- nium ion and precipitation of selenium
onto insulin/chi- tosan complex. The produced selenium Insulin, insulin-like growth factor (IGF-1)
nanoparticles have good insulin encapsulation efficiency and their receptors
as well as gastric sta- bility. Oral administration of the
developed nanoparticles showed remarkable Insulin, to mediate its pharmacological effects, has to
hypoglycaemia in diabetic and non- diabetic rats. The bind to insulin receptor (IR).[133] IR belongs to a class of
study suggests that selenium can potenti- ate the recep- tors known as tyrosine kinase superfamily which
antidiabetic effect of insulin and serve as a new oral comprises IR as well as insulin-like growth factor-1
nanocarrier for the hormone. The study also concluded receptor (IGF- 1R).[134] IR and IGF-1R show significant
that insulin-loaded selenium nanoparticles could have structure homol- ogy, and their configurations are
the almost identical.[134–136]

Figure 5 Simplified diagram of insulin and insulin-like growth factor-1 (IGF-1) signalling pathways. In summary, both are critical to cell health
and survival, and there is significant overlap between the two. PI3K, phosphatidyl inositol 3 kinase, Akt; PkB, protein kinase B; Ras, rat sarcoma
protein; MAPK, mitogen-activated protein kinase. [Colour figure can be viewed at wileyonlinelibrary.com]

208 © 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213
Ahmed Gedawy et al. Oral delivery of insulin

IR and IGF-1R are located on the cell surface, and Insulin binding to IR has been reported to stimulate
both exhibit heterotetrameric architecture.[134–136] Each colon cancer cells through the PI3K/Akt pathway. [139]
receptor is composed of two extracellular ligand binding Regardless of insulin type or origin, a potential mecha-
a-subunits and two transmembrane b-subunits linked by nism of insulin mitogenicity has been explained in a con-
disulphide bonds.[134–136] Another hybrid receptor has dition of hyperinsulinaemia associated with insulin
been identified (IR/IGF-1R) which is composed of one resistance.[140] This unique effect of insulin is mediated
a-subunit and one b-subunit from IR which through IR and not thorough IGF-1 receptor.[140] This
heterodimerize with similar complementary units from mechanism is mediated via activation of
IGF-1R.[134] farnesyltransferase and increases the membrane-anchored
IR/IGF-1R, the hybrid receptor, exhibits higher farnesylated Ras for further activation by insulin as well
affinity towards IGF-1 than insulin and tends to mediate as other growth fac- tors.[140] Another possible, yet
IGF-1 mitogenic responses.[134] Insulin-like growth indirect mitogenic effect of insulin could be achieved by
factors (IGF-1 and IGF-2) are polypeptide ligands that the stimulatory effect of insu- lin on pituitary gland to
share fun- damental structure homology with pro- produce growth hormone which directly governs IGF-1
insulin, and their primary function is to stimulate cell production by liver.[133,136] Long- term use of oral
growth, cell differenti- ation, cell proliferation and cell insulin for a chronic disease is still ques- tionable, and
survival.[133] GIT, liver and other tissues of the body being a growth promoter, oral administra- tion of
synthesize and secrete IGF-1 and IGF-2, and insulin has the potential for inducing mitogenic changes
interestingly, the GIT itself is considered as potential in the gut mucosa which has raised concerns in some
target organ of their mitogenic actions. [136] The close scientists.[66,113]
propinquity between insulin and IGF-1 as possi- ble
ligands and IR and IGF-1 receptor enables insulin and Conclusion
IGF-1 to bind to each other’s receptor and not only to
their cognate receptors. [135] Insulin can potentially bind Non-invasive hormone and peptide administration has
to IGF-1R as well as IGF-2R with much lower affinity. been an interesting challenge for a long time in the
On the other hand, IGF-1 is able to bind to both IGF-1R pharma- ceutical drug delivery field. The limited
and IGF2R and weekly to IR. However, IGF-2 can only administration routes and low oral bioavailability of
bind to its receptor and IGF-1R with no affinity insulin have attracted many scientists since 1922 to
towards the IR.[133] IR predominantly serves in tackle and improve more physiological and non-invasive
expressing the meta- bolic intracellular pathways while insulin delivery options such as buccal, nasal and ocular
IGF-1R primarily initi- ates cell growth.[134] delivery routes. Oral deliv- ery of pharmaceuticals
It has been reported that some mitogenic effects have especially hormones is of higher preference among all
been mediated through the IR, whereas IGF-1 has other administration routes. The con- venience of
contributed somewhat to this.[136] The growth-promoting administration by patients themselves is a major factor
effect of insulin on cell proliferation and differentiation for consideration. In line with this, physicians and
has been shown to be dose-dependent.[134,137] Human clinical diabetologists favour the oral route in diabetes
IGF-1, on the other hand, increased the sensitization to treatment to ensure patient compliance and to achieve
insulin and has improved fast- ing and postprandial the therapeutic targets.
glycaemia in some clinical studies on type 2 diabetic There is a vast body of information in the literature
patients.[134] So, the ligand binding (either insulin or out- lining different techniques and strategies to
IGF-1) to IR or IGF-1R would potentially activate trans- improve oral insulin bioavailability, including proof-
autophosphorylation and the same downstream sig- of-concept studies indicating the potential feasibility of
nalling pathways for cell proliferation, cell successful oral insulin administration. The initial
differentiation, anti-apoptotic effect as well as promising results obtained from these studies have
metabolism.[134,135] been an impetus to undergo phase I and II clinical
Activation of either IR or IGF-1 receptor triggers a trials of some oral insulin products. The intro- duction
series of signalling transduction cascade pathways of some innovative approaches such as mucoadhe- sive
which can gen- erally be categorized into: polymers, absorption enhancers, protease inhibitors as
A Rat sarcoma protein (Ras)/mitogen-activated protein well as particulate carrier systems has boosted the
kinase (MAPK) pathway which significantly scope of research in delivering insulin orally by
mediates mitogenic action and gene expression [133,138] counteracting the naturally existing hurdles and harsh
or conditions of the GIT. Funding, on the other hand, has
B Phosphatidyl inositol 3 kinase (PI3K)/protein always been a paramount limitation to obtain thorough
kinase B (Akt), a pathway that broadly mediates pharmacokinetic and phar- macodynamic data in
insulin metabolic effects (glucose homeostasis) in animals and humans and possible long-term side-
addition to cell migra- tion and anti-apoptotic effects of the newly introduced oral insulin candidates.
action[133,138,139] (Figure 5).

© 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213 209
Oral delivery of insulin Ahmed Gedawy et al.

References nanoparticles vs. subcutaneous 26. Balcerzak SP et al. Surface structure


injec- tion. Biomaterials 2010; 31: of intestinal epithelium. Gastroen-
1. Madhav M. Long-awaited dream of 6849– terology 1970; 58: 49–55.
oral insulin: where did we reach? 6858. 27. Cheng H, Leblond CP. Origin,
Asian J Pharm Clin Res 2011; 4: 16– 14. Arbit E, Kidron M. Oral insulin: the differ- entiation and renewal of the
21. rationale for this approach and cur- four main epithelial cell types in the
2. Chin RL et al. Gas gangrene from rent developments. J Diabetes Sci mouse small intestine. V. Unitarian
subcutaneous insulin administration. Technol 2009; 3: 562–567. theory of the origin of the four
Am J Emerg Med 1993; 11: 622–625. 15. Groop LC et al. Glucose and free epithelial cell types. Am J Anat
3. Saaddine JB et al. Improvements in fatty acid metabolism in non- 1974; 141: 537–561.
diabetes processes of care and inter- insulin- dependent diabetes mellitus. 28. Cornes JS. Number, size, and
mediate outcomes: United States, Evidence for multiple sites of insulin distribu- tion of Peyer’s patches in
1988–2002. Ann Intern Med 2006; resistance. J Clin Investig 1989; 84: the human small intestine. Gut
144: 465. 205. 1965; 6: 225.
16. Rizza RA et al. Dose-response 29. Hiroshi K, Satoshi F. Nalt- versus
4. Morris AD et al. Adherence to peyer’s-patch-mediated mucosal
insu- lin treatment, glycaemic charac- teristics for effects of insulin
on pro- duction and utilization of immunity. Nat Rev Immunol 2004; 4:
control, and ketoacidosis in
insulin-dependent diabetes glucose in man. Am J Physiol 1981; 699.
240: E630. 30. Madara J. Loosening tight junctions.
mellitus. Lancet 1997; 350: 1505–
17. Cherrington A et al. Physiological Lessons from the intestine. J Clin
1510.
consequences of phasic insulin Invest 1989; 83: 1089–1094.
5. Zijlstra E et al. Oral insulin
reloaded: a structured approach. J release in the normal animal. Dia- 31. Salama NN et al. Tight junction
betes 2002; 51: S103–S108. modulation and its relationship to
Diabetes Sci Technol 2014; 8: 458–
465. 18. Wang H et al. Oral administration of drug delivery. Adv Drug Deliv Rev
insulin to female nonobese diabetic 2006; 58: 15–28.
6. Satake S et al. Direct and indirect
effects of insulin on glucose uptake mice inhibited diabetes and induced 32. Larhed A et al. The influence of
Fas ligand expression on islets of intestinal mucus components on the
and storage by the liver. Diabetes
2002; 51: 1663–1671. Langerhans. Chin Med J 2000; 113: diffusion of drugs. Pharm Res 1998;
433–436. 15: 66–71.
7. Matteucci E et al. Insulin adminis-
tration: present strategies and future 19. Wajchenberg BL. Β-cell failure in 33. Macadam A. The effect of gastro-
dia- betes and preservation by clinical intestinal mucus on drug absorption.
directions for a noninvasive
(possibly more physiological) treat- ment. Endocr Rev 2007; 28: Adv Drug Deliv Rev 1993; 11: 201–
187–218. 220.
delivery. Drug Des Dev Ther 2015;
20. Stratton IM et al. Association of gly- 34. Cone RA. Barrier properties of
9: 3109–3118.
8. Lenhard MJ, Reeves GD. caemia with macrovascular and mucus. Adv Drug Deliv Rev 2009;
Continuous subcutaneous insulin microvascular complications of type 61: 75–85.
infusion: a comprehensive review of 2 diabetes (UKPDS 35): prospective 35. Camenisch G et al. Estimation of
insulin pump therapy. Arch Intern observational study. BMJ 2000; permeability by passive diffusion
Med 2001; 161: 2293–2300. 321: 405–412. through Caco-2 cell monolayers
21. Bergerot I et al. Insulin B-chain using the drugs’ lipophilicity and
9. Kordonouri O et al. Treatment of
reac- tive CD 4+ regulatory T-cells molecular weight. Eur J Pharm Sci
type 1 diabetes in children and ado-
induced 1998; 6: 313–319.
lescents using modern insulin
pumps. Diabetes Res Clin Pract by oral insulin treatment protect 36. Moussa BA et al. A validated RP-
from type 1 diabetes by blocking the HPLC method for the
2011; 93: S118–S124.
cytokine secretion and pancreatic determination of recombinant
10. Sameer N. Novel noninvasive tech-
infiltration of diabetogenic effector human insulin in bulk and
niques in management of diabetes.
T-cells. Diabetes 1999; 48: 1720– pharmaceutical dosage form. J
Asian J Pharm 2014; 8: 141–160.
1729. Chem 2010; 7: S449–S457.
11. Hoffman A, Ziv E. Pharmacokinetic
22. Fasano A. Innovative strategies for 37. Evans DF et al. Measurement of
considerations of new insulin formu-
the oral delivery of drugs and pep- gas- trointestinal pH profiles in
lations and routes of administration.
tides. Trends Biotechnol 1998; 16: normal ambulant human subjects.
Clin Pharmacokinet 1997; 33: 285–
152–157. Gut 1988; 29: 1035.
301.
12. Owens D. New horizons-alternative 23. Pauletti GM et al. Structural 38. Sood A, Panchagnula R. Peroral
routes for insulin therapy. Nat Rev require- ments for intestinal route: an opportunity for protein
Drug Discov 2002; 1: 529–540. absorption of peptide drugs. J and peptide drug delivery. Chem
13. Sonaje K et al. Biodistribution, Control Release 1996; 41: 3–17. Rev 2001; 101: 3275–3303.
phar- macodynamics and 24. Kalra S et al. Oral insulin. Diabetol 39. Langguth P et al. The challenge of
pharmacokinetics of insulin Metab Syndr 2010; 2: 66. proteolytic enzymes in intestinal
analogues in a rat model: oral 25. Walker RI, Owen RL. Intestinal bar-
delivery using pH-responsive riers to bacteria and their toxins.
Annu Rev Med 1990; 41: 393.

210 © 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213
Ahmed Gedawy et al. Oral delivery of insulin

peptide delivery. J Control Release 55. Heinemann L, Jacques Y. Oral insu- 68. Agarwal V et al. Oral delivery of
1997; 46: 39–57. lin and buccal insulin: a critical reap- pro- teins: effect of chicken and
40. Schilling R, Mitra A. Degradation of praisal. J Diabetes Sci Technol 2009; duck ovo- mucoid on the stability of
insulin by trypsin and alpha-chymo- 3: 568–584. insulin in the presence of a-
trypsin. Pharm Res 1991; 8: 721– 56. Maher S et al. Formulation strate- chymotrypsin and trypsin. Pharm
727. gies to improve oral peptide deliv- Pharmacol Commun 2000; 6: 223–
41. Chang L-L, Bai J. Evidence for the ery. Pharm Pat Anal 2014; 3: 313– 227.
existence of insulin-degrading 336. 69. Aungst BJ. Intestinal permeation
enzyme on the brush-border mem- 57. Kidron M et al. A novel per-oral enhancers. J Pharm Sci 2000; 89: 429–
branes of rat enterocytes. Pharm Res insulin formulation: proof of 442.
1996; 13: 801–803. concept study in non-diabetic 70. Brayden DJ, Mrsny RJ. Oral peptide
42. Binder C et al. Insulin pharmacoki- subjects. Diabet Med 2004; 21: 354. delivery: prioritizing the leading
netics. Diabetes Care 1984; 7: 188. 58. Chalasani KB et al. Effective oral tech- nologies. Ther Deliv 2011; 2:
43. Juan Pablo M et al. Anti-diabetic delivery of insulin in animal models 1567.
activity of insulin-degrading enzyme using vitamin B12-coated dextran 71. Fasano A, Uzzau S. Modulation of
inhibitors mediated by multiple hor- nanoparticles. J Control Release intestinal tight junctions by zonula
2007; 122: 141–150. occludens toxin permits enteral
mones. Nature 2014; 511: 94–98.
59. Khafagy E-S, Morishita M. Oral administration of insulin and other
44. Carino GP, Mathiowitz E. Oral insu- macromolecules in an animal model.
lin delivery. Adv Drug Deliv Rev bio- drug delivery using cell-
penetrating peptide. Adv Drug J Clin Investig 1997; 99: 1158.
1999; 35: 249–257. 72. Touitou E, Rubinstein A. Targeted
45. Michael G, Isabel G-O. Challenges Deliv Rev 2012; 64: 531–539.
60. Tréhin R, Merkle HP. Chances enteral delivery of insulin to rats. Int
for the oral delivery of
and pitfalls of cell penetrating J Pharm 1986; 30: 95–99.
macromolecules. Nat Rev Drug
peptides for cellular drug delivery. 73. Li J et al. In vitro evaluation of dis-
Discov 2003; 2: 289.
Eur J Pharm Biopharm 2004; 58: solution behavior for a colon-
46. Agarwal V, Khan MA. Current status
209–223. specific drug delivery system
of the oral delivery of insulin. Pharm (CODESTM) in multi-pH media
61. Herce H, Garcia A. Cell penetrating
Technol 2001; 25: 76–90. using United States pharmacopeia
peptides: how do they do it? J Biol
47. Mahato R et al. Emerging trends in apparatus II and III. AAPS
Phys 2007; 33: 345–356.
oral delivery of peptide and protein PharmSciTech 2002; 3: 59–67.
62. Liang JF, Yang VC. Insulin-cell
drugs. Crit Rev Ther Drug Carr Syst pene- trating peptide hybrids with 74. Katsuma M et al. Effects of absorp-
2003; 20: 153–214. improved intestinal absorption effi- tion promoters on insulin absorption
48. Roberts MJ et al. Chemistry for pep- ciency. Biochem Biophys Res Comm through colon-targeted delivery. Int
tide and protein pegylation. Adv 2005; 335: 734–738. J Pharm 2006; 307: 156–162.
Drug Deliv Rev 2002; 54: 459–476. 63. Van Der Lubben IM et al. Chitosan 75. Diabetology Ltd. CapsulinTM;
49. Hinds KD, Kim SW. Effects of peg for mucosal vaccination. Adv Drug 2017 Available from:
conjugation on insulin properties. Adv Deliv Rev 2001; 52: 139–144. http://www.diabetol
Drug Deliv Rev 2002; 54: 505–530. 64. Pappo J, Ermak T. Uptake and ogy.co.uk/projects/capsulin/
50. Greenwald RB et al. Effective drug translocation of fluorescent latex-par- (accessed 02 August 2017).
delivery by pegylated drug conjugates. ticles by rabbit Peyer’s patch follicle 76. Soares S et al. Novel non-invasive
Adv Drug Deliv Rev 2003; 55: 217– epithelium – a quantitative model methods of insulin delivery. Expert
250. for M cell uptake. Clin Exp Immunol Opin Drug Deliv 2012; 9: 1539–
51. Clement S et al. Oral insulin product 1989; 76: 144–148. 1558.
hexyl-insulin monoconjugate 2 65. Pan Y et al. Bioadhesive polysaccha- 77. Luzio SD et al. The glucose lowering
(HIM2) in type 1 diabetes mellitus: ride in protein delivery system: chi- effect of an oral insulin (capsulin)
the glucose stabilization effects of tosan nanoparticles improve the during an isoglycaemic clamp study
HIM2. Diabetes Technol Ther 2002; intestinal absorption of insulin in in persons with type 2 diabetes. Dia-
4: 459. vivo. Int J Pharm 2002; 249: 139– betes Obes Metab 2010; 12: 82–87.
52. Owens DR et al. Alternative routes 147. 78. Miriam K et al. Methods and com-
of insulin delivery. Diabet Med 66. Iyer H et al. Oral insulin – a review positions for oral administration of
2003; 20: 886–898. of current status. Diabetes Obes proteins. United States patent
53. Morishita M, Peppas NA. Is the oral Metab 2010; 12: 179–185. US20110142800 A1. 2011.
route possible for peptide and pro- 67. Yamamoto A et al. Effects of 79. Oramed Pharmaceuticals. Oral insu-
tein drug delivery? Drug Discov various protease inhibitors on the lin delivery pipeline; 2017 Available
Today 2006; 11: 905–910. intestinal absorption and from: http://www.oramed.com/pipe
54. Wajcberg E et al. Dose-response degradation of insu- lin in rats. line/ (accessed 02 August 2017).
effect of a single administration of AAPS PharmSciTech 1994; 11: 80. Dorkoosh FA et al. Development
oral hexyl-insulin to monoconjugate 1496–1500. and characterization of a novel
2 in healthy nondiabetic subjects. peroral peptide drug delivery
system. J Con- trol Release 2001; 71:
Diabetes Care 2004; 27: 2868–2873.
307–318.

© 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213 211
Oral delivery of insulin Ahmed Gedawy et al.

81. Yin L et al. Beneficial properties for cells. Cell Tissue Res 1995; 279: 433– recent advances in self-
insulin absorption using superporous 436. nanoemulsify- ing drug delivery
hydrogel containing interpenetrating 93. Pan Y et al. Relationship between drug systems (SNEDDS). Expert Opin
polymer network as oral delivery effects and particle size of insulin- Drug Deliv 2016; 00: 1–
vehicles. Int J Pharm 2008; 350: 220– loaded bioadhesive microspheres. Acta 16.
229. 105. Chime SA et al. Nanoemulsions –
Pharmacol Sin 2002; 23: 1051–1056.
82. Yin L et al. Polymer integrity advances in formulation, characteri-
94. Ubaidulla U et al. Development and
related in-vivo evaluation of insulin-loaded zation and applications in drug
absorption mechanism of super- chitosan phthalate microspheres for delivery, application of nanotechnol-
porous hydrogel containing oral delivery. J Pharm Pharmacol ogy in drug delivery. InTech 2014;
interpen- etrating polymer networks https://doi.org/10.5772/58673.
2007; 59: 1345–1351.
for oral delivery of insulin. 106. Rao SV, Shao J. Self-
95. Toorisaka E et al. An enteric-coated
Biomaterials 2010; 31: 3347–3356. nanoemulsifying drug delivery
dry emulsion formulation for oral
83. Smart JD. The basics and systems (SNEDDS) for oral delivery
underlying mechanisms of insulin delivery. J Control Release of protein drugs: I. For- mulation
mucoadhesion. Adv Drug Deliv Rev 2005; 107: 91–96.
development. Int J Pharm 2008;
2005; 57: 1556–1568. 96. Sharma G et al. Microemulsions for 362: 2–9.
84. Lehr C-M. Lectin-mediated drug oral delivery of insulin: design,
devel- opment and evaluation in 107. Sakloetsakun D et al. Combining
delivery: the second generation of two technologies: multifunctional
bioadhesives. J Control Release streptozo- tocin induced diabetic
polymers and self-nanoemulsifying
2000; 65: 19–29. rats. Eur J Pharm Biopharm 2010;
drug delivery system (SNEDDS) for
85. Bernkop-Schnu € rch A. Thiomers: 76: 159–169.
oral insulin administration. Int J
a new generation of mucoadhesive 97. Li P et al. Oral delivery of peptides
Biol Macromol 2013; 61: 363–372.
polymers. Adv Drug Deliv Rev and proteins using lipid-based drug
delivery systems. Expert Opin Drug 108. Zhang QH et al. The in vitro and
2005; 57: 1569–1582. in vivo study on self-
86. Krauland AH et al. Oral insulin Deliv 2012; 9: 1289–1304.
nanoemulsifying drug delivery
delivery: the potential of thiolated 98. Patel HM, Ryman BE. Oral system (SNEDDS) based on insulin-
chitosan-insulin tablets on non-dia- adminis- tration of insulin by
phospholipid complex. J Biomed
betic rats. J Control Release 2004; encapsulation within liposomes.
FEBS Lett 1976; 62: 60–63. Nanotechnol 2012; 8: 90–97.
95: 547–555. 109. Karamanidou T et al. Effective
87. Pereira De Sousa I et al. Insulin 99. Geho WB et al. Hepatic-directed
vesicle insulin: a review of formula- incor- poration of insulin in mucus
loaded mucus permeating nanoparti- perme- ating self-nanoemulsifying
cles: addressing the surface tion development and preclinical
evaluation. J Diabetes Sci Technol drug delivery systems. Eur J Pharm
character- istics as feature to
2009; 3: 1451. Bio- pharm 2015; 97: 223–229.
improve mucus permeation. Int J
100. Blair Geho W et al. Insulin 110. Prego C et al. Transmucosal macro-
Pharm 2016; 500: 236–244.
therapies for the treatment of molecular drug delivery. J Control
88. Huckaby JT, Lai SK. Pegylation for
enhancing nanoparticle diffusion in diabetes, diabetes related ailments, Release 2005; 101: 151–162.
mucus. Adv Drug Deliv Rev 2017; and/or diseases or conditions other 111. Fonte P et al. Oral insulin delivery:
https://doi.org/10.1016/j.addr.2017.0 than diabetes or dia- betes related how far are we? J Diabetes Sci Tech-
8. 010. ailments. United States patent nol 2013; 7: 520.
US20120035105 A1. 2012. 112. Sonia TA, Sharma CP. An overview
89. Liu M et al. Efficient mucus perme-
101. Almeida AJ, Souto E. Solid lipid of natural polymers for oral insulin
ation and tight junction opening by nanoparticles as a drug delivery sys-
dissociable “mucus-inert” agent delivery. Drug Discov Today 2012;
tem for peptides and proteins. Adv
coated trimethyl chitosan nanoparti- 17: 784–792.
cles for oral insulin delivery. J Drug Deliv Rev 2007; 59: 478–490. 113. Chen M-C et al. A review of the
Control Release 2016; 222(Suppl. 102. Mu € ller RH et al. Solid lipid pro- spects for polymeric
C): 67–77. nanopar- ticles (SLN) for controlled nanoparticle platforms in oral insulin
drug delivery – a review of the state delivery. Biomaterials 2011; 32:
90. Crdenas-Bailn F et al.
Microencapsula- tion techniques to of the art. Eur J Pharm Biopharm 9826–9838.
develop formulations of insulin for 2000; 50: 161–177. 114. Hillaireau H, Couvreur P. Nanocarri-
oral delivery: a review. J 103. Garcia-Fuentes M et al. Design of ers’ entry into the cell: relevance to
Microencapsul 2013; 30: 409–424. lipid nanoparticles for the oral deliv- drug delivery. Cell Mol Life Sci 2009;
ery of hydrophilic macromolecules. 66: 2873–2896.
91. Hamman J et al. Oral delivery of
Colloid Surf B Biointerfaces 2003; 115. Florence AT. Nanoparticle uptake by
peptide drugs. BioDrugs 2005; 19: 27: the oral route: fulfilling its potential?
165–177.
159–168. Drug Discov Today Technol 2005; 2:
92. Ermak T et al. Uptake and transport
104. Rehman FU et al. From nanoemul- 75–81.
of copolymer biodegradable micro-
sions to self-nanoemulsions, with 116. Joshi HM et al. Gold nanoparticles
spheres by rabbit Peyer’s patch M
as carriers for efficient transmucosal

212 © 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213
Ahmed Gedawy et al. Oral delivery of insulin

insulin delivery. Langmuir 2006; 22: 126. Zhang P et al. Goblet cell targeting 133. Wilcox G. Insulin and insulin resis-
300–305. nanoparticle containing drug-loaded tance. Clin Biochem Rev 2005; 26:
117. Sharif H et al. Nanoparticles and micelle cores for oral delivery of 19.
toxicity in therapeutic delivery: the insu- lin. Int J Pharm 2015; 496: 134. Tennagels N, Werner U. The meta-
993–1005. bolic and mitogenic properties of
ongoing debate. Ther Deliv 2011; 2:
127. Boushra M et al. Methocel-lipid basal insulin analogues. Arch
125–132.
hybrid nanocarrier for efficient oral Physiol Biochem 2013; 119: 1–14.
118. Nel A et al. Toxic potential of mate-
insulin delivery. J Pharm Sci 2016; 135. Weinstein D et al. Insulin receptor
rials at the nanolevel. Science 2006;
105: 1733–1740. compensates for IGF1R inhibition
311: 622–627.
128. Sahoo P et al. Optimization of pH- and directly induces mitogenic
119. Panariti A et al. The effect of activ- ity in prostate cancer cells.
responsive carboxymethylated iota-
nanopar- ticle uptake on cellular Endocr Connect 2014; 3: 24–35.
carrageenan/chitosan nanoparticles
behavior: dis- 136. Kuemmerle JF. Insulin-like growth
for oral insulin delivery using
rupting or enabling functions? response surface methodology. factors in the gastrointestinal tract
Nanotechnol Sci Appl 2012; 5: 87. React Funct Polym 2017; and liver. Endocrinol Metab Clin
120. Krpetić Ž et al. Nanomaterials: 119(Suppl. C): 145–155. North Am 2012; 41: 409–423.
impact on cells and cell organelles. 129. Ji N et al. Binary and tertiary com- 137. Weinstein D et al. Insulin analogues
Adv Exp Med Biol 2014; 811: 135– plex based on short-chain glucan display IGF-I-like mitogenic and
136. and proanthocyanidins for oral anti-apoptotic activities in cultured
121. Buzea C et al. Nanomaterials and insulin delivery. J Agric Food Chem cancer cells. Diabetes Metab Res
nanoparticles: sources and toxicity. 2017; 65: 8866–8874. Rev 2009; 25: 41–49.
Biointerphases 2007; 2: MR17– 130. Deng W et al. Selenium nanoparti- 138. Yu H, Rohan T. Role of the insulin-
MR71. cles as versatile carriers for oral like growth factor family in cancer
122. Panyam J, Labhasetwar V. delivery of insulin: insight into the development and progression. J Natl
Biodegrad- able nanoparticles for synergic antidiabetic effect and Cancer Inst 2000; 92: 1472–1489.
drug and gene delivery to cells and mechanism. Nanomed Nanotechnol 139. Wang Y et al. Mitogenic and anti-
tissue. Adv Drug Deliv Rev 2003; 55: Biol Med 2017; 13: 1965–1974. apoptotic effects of insulin in
329–347.
131. Bhattacharyya A et al. Preparation endome- trial cancer are
123. Unfried K et al. Cellular responses of polyurethane–alginate/chitosan phosphatidylinositol 3-kinase/Akt
to nanoparticles: target structures core shell nanoparticles for the dependent. Gynecol Oncol 2012; 125:
and mechanisms. Nanotoxicology purpose of oral insulin delivery. Eur 734–741.
2007; 1: 52–71. 140. Draznin B. Mitogenic action of insu-
Polymer J 2017; 92(Suppl. C): 294–
124. Banerjee A et al. Intestinal mucoad- 313. lin: friend, foe or ‘frenemy’? Dia-
hesive devices for oral delivery of betologia 2010; 53: 229.
132. Sun S et al. Multifunctional
insulin. Bioeng Transl Med 2016; 1: composite microcapsules for oral
338–346.
delivery of insu- lin. Int J Mol Sci
125. Banerjee A et al. Intestinal micro- 2017; 18: 54.
patches for oral insulin delivery. J
Drug Target 2017; 25: 608–615.

© 2017 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 197–213 213

You might also like