Sei sulla pagina 1di 21

Journal of Controlled Release 178 (2014) 25–45

Contents lists available at ScienceDirect

Journal of Controlled Release


journal homepage: www.elsevier.com/locate/jconrel

Review

Advanced progress of microencapsulation technologies: In vivo and


in vitro models for studying oral and transdermal drug deliveries
P.L. Lam a,⁎, R. Gambari b
a
State Key Laboratory of Chirosciences, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
b
Centre of Biotechnology, Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy

a r t i c l e i n f o a b s t r a c t

Article history: This review provides an overall discussion of microencapsulation systems for both oral and transdermal drug
Received 30 August 2013 deliveries. Clinically, many drugs, especially proteins and peptides, are susceptible to the gastrointestinal tract
Accepted 17 December 2013 and the first-pass metabolism after oral administration while some drugs exhibit low skin permeability through
Available online 10 January 2014
transdermal delivery route. Medicated microcapsules as oral and transdermal drug delivery vehicles are believed
to offer an extended drug effect at a relatively low dose and provide a better patient compliance. The polymeric
Keywords:
Microencapsulation
microcapsules can be produced by different microencapsulation methods and the drug microencapsulation tech-
Oral drug delivery nology provides the quality preservation for drug stabilization. The release of the entrapped drug is controlled
Transdermal drug delivery and prolonged for specific usages. Some recent studies have focused on the evaluation of drug containing micro-
In vivo study capsules on potential biological and therapeutic applications. For the oral delivery, in vivo animal models were
Skin delivery model used for evaluating possible treatment effects of drug containing microcapsules. For the transdermal drug deliv-
ery, skin delivery models were introduced to investigate the potential skin delivery of medicated microcapsules.
Finally, the challenges and limitations of drug microencapsulation in real life are discussed and the commercially
available drug formulations using microencapsulation technology for oral and transdermal applications are
shown.
© 2013 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
2. Challenges to oral and transdermal drug deliveries of conventional routes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
2.1. Oral delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
2.2. Transdermal delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
3. Microencapsulation for drug deliveries . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
4. Microencapsulation techniques for drug entrapment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
4.1. Chemical microencapsulation processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
4.2. Physicochemical microencapsulation processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
4.3. Mechanical microencapsulation processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
5. Stabilization of drugs using microencapsulation technology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
6. Drug release mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
6.1. Diffusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
6.2. Swelling and dissolution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
6.3. Erodible and degradation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
6.4. Osmosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
7. Recent advances in microencapsulated oral and transdermal drug delivery systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
7.1. Natural polymer-based systems for drug microencapsulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
7.1.1. Chitosan-based systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
7.1.2. Gelatin-based systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
7.1.3. Alginate-based systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
7.1.4. Cellulose-based systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37

⁎ Corresponding author.
E-mail address: tcstacey@inet.polyu.edu.hk (P.L. Lam).

0168-3659/$ – see front matter © 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.jconrel.2013.12.028
26 P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45

7.2. Synthetic polymer-based systems for drug microencapsulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37


7.2.1. Poly(L-lactic acid) (PLA)- and poly(lactic-coglycolic acid) (PLGA)-based systems . . . . . . . . . . . . . . . . . . . . . . . . 37
7.2.2. Poly (vinyl alcohol) (PVA)-based systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
7.2.3. Poly(ɛ-caprolactone) (PCL)-based systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
7.2.4. Poly(methyl methacrylate) (PMAA)-based systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
7.3. Multiple microemulsion systems for drug microencapsulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
8. Challenges of drug microencapsulation in real life . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
9. Commercially available microencapsulated pharmaceuticals for oral and transdermal drug deliveries . . . . . . . . . . . . . . . . . . . . . . . 40
10. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40

1. Introduction manner, for example, the core ingredients are controlled to be released
all at once or moderately and gradually. Microcapsules are generally
Advanced technologies in drug delivery turn the challenges into produced within the size ranging from 1 to 1000 μm for enabling the ef-
opportunities of drug deliveries. With the progressive developments ficiency of the controlled release profile of core active substances and
and explorations in technologies, traditional drug delivery routes extending the shelf life of entrapped substances [10–22]. Microencapsu-
are being supplemented by sophisticated and versatile approaches lation has been widely applied to the design and development of drug
of drug deliveries. Recently, intensive studies have focused on the inno- delivery systems as microcapsules could protect the drugs from degra-
vative technologies to tackle the restrictions of conventional drug deliv- dation and prevent the body from the potential toxicity induced by
ery systems. It is highly challenging to develop biopharmaceuticals or drugs [23]. This review highlights a comprehensive summary of chal-
biomedical drugs with an optimal bioavailability and biostability as lenges to conventional oral and transdermal drug delivery routes, micro-
well as a maximum therapeutic efficiency. The desired oral dosages encapsulation techniques for drug deliveries, drug stabilization using
should protect the drugs under unstable biological environments in- microencapsulation, drug release mechanism of drug loaded microcap-
cluding drug degradation induced by the gastrointestinal tract and sules, and advances in oral and transdermal drug deliveries using micro-
first-pass liver effects after oral administration before reaching the encapsulation technologies to overcome these challenges. The challenges
targeted sites, and maximize the drug uptake and absorption in the cel- and limitations of drug microencapsulation in real life and the commer-
lular intestinal regions [1]. The development of topical formulations cially available pharmaceuticals using microencapsulation technology
should enhance the drug absorption, alleviate the skin disorder by in- for oral and transdermal drug deliveries are shown.
creasing the skin hydration to overcome the low skin permeability,
and promote a prolonged delivery time to the targeted skin sites [2]. 2. Challenges to oral and transdermal drug deliveries of
In fact, many drugs, such as anticancer drugs with 5–20% oral bioavail- conventional routes
ability [3], proteins and peptides with less than 1–2% oral bioavailability
[4], are limited in their low bioavailability when they are taken orally 2.1. Oral delivery
while some patients are found to have immediate and delayed allergy
and site irritation to intravenous injection [5]. As a result, many strate- Oral delivery is considered as an attractive and preferable route for
gies have been developed for oral and transdermal drug delivery systems drug administration as oral administration provides some advantages
using microencapsulation technologies because these provide some pos- for people, such as ease and full control of drug administration by pa-
sible advantages for drug deliveries over conventional routes. Micro- tients, together with a high degree of flexibility on dosages [24]. Previous
particulate systems for oral and transdermal drug deliveries are speculat- studies reported that almost all patients preferred oral administration
ed to improve the drawbacks in the traditional drug dosage forms, and route of anti-cancer drugs instead of intravenous route. Approximately
offer an alternative administration route to enhance the release profile 80% of patients chose to have an oral chemotherapy treatment for breast
and improve the bioavailability of drugs. The efficacy of many drugs in cancer while less than 3% of them favored parenteral route [25]. In anoth-
conventional dosage forms, including film coated tablets and hard or er study, almost 90% of patients preferred oral chemotherapy for colon
soft gelatin capsules, is often restricted by their limitations to be deliv- cancer to intravenous treatment [26]. More than 60% of commercially
ered to targeted body sites as the drugs may be degraded by the acidic available drug dosage forms with small molecules are orally adminis-
gastric fluid and this induces irritation to stomach [6]. Therefore, tered [27]. However, many conventional drugs exhibit poor bioavailabil-
searching biotechnologies to offer the desired excipients to accomplish ity when they are taken orally. In oral drug delivery route, impermeable
the targeted drug delivery is particularly important. gastrointestinal epithelium behaves as a physical barrier while peptidase
Medicated-polymeric microcapsules as oral and topical drug deliv- induced enzymatic degradation acts as a biochemical barrier. The gastro-
ery vehicles are believed to minimize the inconveniences and risks dur- intestinal tract, with the total length of approximately 20 ft, is mainly di-
ing oral and intravenous administrations, promote the extension of vided into two parts: the upper part involves mouth, pharynx, esophagus
drug effect at a relatively low dose, and offer the ease of drug adminis- and stomach; and the lower part includes small intestine (duodenum,
tration, resulting in a better patient compliance [7–10]. Microencapsula- jejunum and ileum), large intestine (cecum, colon and rectum) and
tion, as a micro-packaging technique, involves the manufacture of anus [28]. More than 90% of nutrients including proteins, minerals, carbo-
microcapsules or microspheres by surrounding the small particles of hydrates, fats, vitamins and water are absorbed in the small intestine
solids, droplets of liquids or dispersions of solids in liquids with the [28], followed by entering the lymphatic circulation or bloodstream to
polymeric matrix or shell to give small capsules a variety of applications. the first-pass metabolism (liver). However, when the drug is absorbed
An encapsulated reservoir of core active ingredients is under the con- through the bloodstream to the first-pass metabolism, the hepatic
trolled release to supply valuable and useful effects for specific end- enzymes usually degrade the drug, resulting in low bioavailability and
uses. The wall material is able to prevent the core active ingredients limited efficacy of the orally administered dosage form. In fact, many
from the damages of external environments such as acidity, alkalinity, drugs, especially protein and peptide-based medicines, exhibit the oral
evaporation, heat, oxidization, light or moisture. Micro-particulate car- bioavailability below 1%. These drugs are difficult to pass through muco-
riers enable the controlled release of core active substances in a targeted sal surfaces and biological membranes, and they are being inactivated
P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45 27

because of the stomach juice with strong acid or digestive protease en-
zymes within the gastrointestinal tract. Additionally, drugs are unable
to be effectively absorbed into the systematic circulation due to their
large molecular size, charge issues, and hydrophilic property, resulting
in the tendency to be denaturated and aggregated in the first-pass me-
tabolism [29–32]. Besides the protein and peptides, delivery of anti-
cancer drugs through oral route is also greatly challenged because of
their peculiar physicochemical properties and the physiological barriers
including pre-systemic metabolism and gastrointestinal instability. Oral
bioavailability of some anti-cancer medicines such as doxorubicin, doce-
taxel, paclitaxel and tamoxifen only ranges from 5 to 20% [3,33–36].
Fig. 1. Schematic diagrams of (a) an o/w system and (b) a w/o system.
2.2. Transdermal delivery

Transdermal delivery is to apply the drugs or medicines to the skin, be applied to the deliveries of hydrophilic agents, including water-
the drawbacks of oral administration such as enzymatic degradation soluble vitamin B and C, antibiotics, and chemotherapeutic drugs (e.g.
and rapid clearance in the gastrointestinal tract or the first-pass metabo- cisplatin).
lism, therefore, can be avoided. Targeted drug delivery to skin sites By using the microencapsulation technology, many benefits can be
alleviates the physical pain and discomfort, and also promotes the found in both oral and transdermal drug deliveries. Microencapsulation
patient convenience and compliance for drug treatments [37–39]. provides a physical barrier to protect the drugs which are liable to
Although the skin offers a relatively large and readily accessible surface external environments such as acidity, alkalinity, evaporation, heat,
area for drug adsorption, skin-targeted drug delivery still presents limita- oxidization, light or moisture before drug release in order to improve
tions. The human skin is an impermeable barrier that provides a strong the stability of drugs [16]. Curcumin, which is susceptible to oxidants,
protection against the external substances including bacteria, fungi, vi- light and heat, was protected by gelatin/porous starch microcapsules to
ruses, allergens, dust and large molecules. The skin is mainly composed avoid damages and degradation during industrial process. The microcap-
of two layers: the underlying layer and the upper basement layer. In sule-stabilized curcumin still preserved the antimicrobial activities
the underlying layer, various types of cells, blood vessels, lymphatics against foodborne pathogens and spoilage microbe, such as Escherichia
and nerves are found in a dense network of connective tissue. In the coli (E. coli), Yersinia enterocolitica (Y. enterocolitica), Staphylococcus aureus
upper basement membrane, more than 90% of stratified keratinocytes (S. aureus), Bacillus subtilis (B. subtilis), Bacillus cereus (B. cereus),
exist, and keratinocytes undergo the cell differentiation and move up- Aspergillus niger (A. niger), Penicillium notatum (P. notatum), and Saccharo-
ward from the stratum basale, through the stratum spinosum and stra- myces cerevisiae (S. cerevisiae) [65]. Ascorbic acid is considered as an anti-
tum granulosum, to the outermost layer, the stratum corneum to oxidant agent to stabilize free radicals that lead over time to degenerative
finally become corneocytes. Corneocytes grow into anucleated and flat- diseases such as cardiovascular diseases and cancer. However, it is highly
tened cells and they are eventually sloughed off [40]. The stratum unstable and easily degraded. Microencapsulated ascorbic acid was syn-
corneum, as an outermost layer of epidermis, provides an extremely ef- thesized by using four coating materials including gel, starch, ethyl cellu-
fective barrier for the control of drug penetration by an incapability of a lose and β-cyclodextrin. The results revealed that microencapsulation
majority of drugs to pass the skin at therapeutic rates and it is also the could avoid the color change, decelerate the release rate and mask
main barrier to spread water out of the skin [37,40–42]. The anucleated, the acid taste of ascorbic acid. Starch and β-cyclodextrin coated micro-
lattened and protein-rich corneocytes of the stratum corneum are thickly capsules could delay the degradation of ascorbic acid when they
packed within the extracellular lipid matrix [40]. Therefore, the major were stored at 38 °C [66]. Taste marking can be performed by using
challenge of transdermal drug delivery is to overcome the strong barrier microencapsulation systems. Many oral medicines or pharmaceuticals
function of the skin as this barrier leads to slow drug penetration rates, usually have an obnoxious taste such as bitterness. MicroMask®
limited drug uptake rate and lack of dosage flexibility or precision. technology incorporates a dry-powder micro-particulate approach to
conceal the unpleasant taste or odor of drugs within a specialized matrix
3. Microencapsulation for drug deliveries in order to deal with the unwilling patients to take medicines [67].
Berberine was entrapped in the hydroxypropylmethylcellulose micropar-
In the 21st century, microencapsulation technology has been ticles and the microparticles were then coated with Eudragit E100 in
extensively applied to drug deliveries. Biodegradable polymeric micro- order to mask the bitter taste of the drug [68]. Microencapsulated drugs
capsules have received great attention as potential drug delivery vehicles or compounds also improve the drawbacks of conventional drug delivery
in consideration of their applications in targeted drug delivery. routes including poor bioavailability and compatibility, low absorption,
Microparticles have already been introduced to drug delivery systems and short half-life. Microencapsulation increases the therapeutic efficien-
with great success. Microcapsule drug carrier systems possess high poten- cy of drugs and minimizes environmental damages to drugs by making
tial for various applications in therapeutic and pharmaceutical fields, such use of the property of the controlled release. The long-term therapeutic
as anti-inflammation [43–45], antibiotics [46–50], anti-tumor [9,51–56], performance could be enhanced by microencapsulated drugs with an ap-
proteins [57–61] and vitamins [62–64]. Biopharmaceuticals can be simply propriate host response in a specific therapeutic purpose, for example,
produced by an oil-in-water (o/w) system or a water-in-oil (w/o) system. chronic disease treatments require a continuous delivery of the drug
Generally, an o/w system refers to the processing emulsion in which the [16]. Protein-based drugs, such as insulin, have poor oral bioavailability
oil is used as a dispersed phase while the water-soluble/hydrophilic ma- and short half-life after administration because they are sensitive to enzy-
terial is employed as a dispersed medium during the microcapsule forma- matic degradation. For the treatment of iron-deficiency chronic anemia,
tion process. However, a w/o system refers to the processing emulsion patients require at least three to four injections per day to achieve the ap-
which contains the water-soluble/hydrophilic material as the dispersed proximately normal glycemia level for a long-term period. This causes
phase and the oil as the dispersed medium during the formation process. discomfort sufferings, inconveniences and low compliance to patients. In-
Fig. 1 shows the schematic diagrams of an o/wsystem and a w/o system. sulin microcapsules were deposited with multi layers of oppositely
For the potential applications, an o/w system could be applied to the charged Fe3+ and dextran sulfate (DS) in order to offer a longer lasting
deliveries of oil-soluble agents, including fat-soluble vitamin A, D, E and performance. The insulin microcapsules greatly improved glucose toler-
K, as well as water-insoluble drugs (e.g. celecoxib). A w/o system could ance from 2 h (free insulin) to even 12 h (insulin microcapsules with
28 P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45

ten bilayers) [69]. Poly(3-hydroxybutyrate-co-3-hydroxyvalerate) mi- materials to work easily. Park et al. reported the poly(butylene succi-
croparticles were developed for the ibuprofen delivery. The drug-loaded nate)/poly(ɛ-caprolactone) (PBS/PCL) microcapsules containing indo-
microcapsules with multilayer shells of chitosan and sodium alginate methacin prepared by emulsion solvent evaporation method [76].
were demonstrated to be more effective to prolong the drug half release Hamishehkar et al. developed insulin-loaded PLGA microcapsules with
to 62 h with a reduced initial burst when compared with the microcap- respirable size using oil in oil emulsification/solvent evaporation method
sules without multilayer coverage, with only 1 h of drug half release [77].
[70]. A long-term delivery of erythropoietin was achieved by alginate-
poly-L-lysine-aginate microencapsulated C2C12 myoblasts. The results 4. Microencapsulation techniques for drug entrapment
showed that the continuous release of erythropoietin could last for
more than 7months [71]. Microencapsulated drug delivery systems pro- Drug microencapsulation can be achieved by using different micro-
vide various release profiles of drugs or compounds in a controlled man- encapsulation techniques. The most commonly used microencapsulation
ner whether the core drug is released all at once or moderately and methods for drug entrapment involve: (i) chemical processes (interfacial
gradually [16]. The release rate of the drug could be affected by the alter- and free radical polymerization methods); (ii) physico-chemical
ation of processing parameters during the microcapsule formation process. processes (coacervation (phase separation) and ionotropic gelation
The release profiles of bovine erythrocyte hemoglobin (Hb) from poly(L- methods); and (iii) mechanical processes (fluid bed coating, solvent
histidine)-chitosan/alginate complex microcapsules prepared with low, evaporation/extraction and spray drying methods). Table 1 summarizes
medium and high molecular weight of chitosan were studied. Microcap- the microencapsulation methods for drug delivery applications, the
sules prepared with low, medium and high molecular weight of chito- particle size and stability of their resulted microcapsules, advantages
san showed the initial burst release of Hb of 5.3%, 14% and 9.4% in the and disadvantages of these methods, as well as examples using these
first 30 min and 14.4%, 26.3%, 20.5% after 2 h, respectively. The Hb re- methods for drug deliveries.
lease reached 84.8%, 71.4%, and 87.3% from microcapsules prepared
with low, medium and high molecular weight of chitosan after 72 h
[72]. The release profiles of phenacetin loaded gelatin-based microcap- 4.1. Chemical microencapsulation processes
sules prepared with different poor solvents, including methanol, etha-
nol, and propanol, were investigated. Microcapsules prepared with Interfacial polymerization refers to the formation of a polymer at the
methanol were fragile and unacceptable. The results showed that the al- interface between two liquid phases. The wall of microcapsules is
teration of poor solvent during microcapsule formation process could formed at or on the surface of a droplet or particle by the reactive mono-
affect the drug release rate. Propanol prepared microcapsules possessed mer polymerization. A multi-functional monomer is dissolved in the liq-
a slower drug release (lower than 34%) than ethanol prepared micro- uid form of core materials, and the mixture is dispersed to a desired
capsules (more than 60%) after 24 h [73]. Microencapsulation also drop size in an aqueous phase involving a dispersing agent and a
achieves an effective drug concentration at the targeted body sites for multi-functional amine. The rapid reaction of polymerization then gen-
an extended period of time, with minimal systemic exposure. The erates the wall shell of microcapsules [16,23,78]. Free radical polymeri-
local cancer therapy could be achieved by the synergistic effect of radi- zation involves an initiator and a monomer. The initiator molecules are
ation and the anticancer drug. The hyaluronic acid/alginate microcap- firstly converted to free radicals by heating, photolysis or electrolysis.
sules released the carboplatin in response to radiation, resulting in a The free radicals then become highly active to obtain electrons
targeted drug delivery to the intratumoral sites. The mice model from the molecules of the monomers. The microparticles are formed
showed that the combination of microencapsulated carboplatin and ra- through the growth of polymer chains as a result of electron transfer be-
diation remarkably increased the intratumoral carboplatin concentra- tween the reactive monomers [79]. Drug loaded microparticles are pro-
tion, and the significant increase in carboplatin concentration was duced by imbibing the dried microparticles in the drug solution [80–82].
maintained for 48 h after irradiation. Administration of microencapsu-
lated carboplatin did not induce a noticeable increase in the blood 4.2. Physicochemical microencapsulation processes
carboplatin level while the free carboplatin highly increased the drug
concentration in blood from 1 to 6 h after administration. The lower Coacervation (phase separation) was initially introduced by
blood carboplatin concentration could reduce the adverse reactions Bungenberg de Jong & Kruyt in 1929. It was defined as partial
and side effects of carboplatin [74]. Microencapsulated drugs reduce desolvation of a homogeneous polymer solution into a polymer-rich
the gastrointestinal irritation, potential toxicity and side effects of the phase (coacervate) and a poor polymer phase (coacervation medium)
original drugs. DepoMed, Inc. has developed the controlled release [83,84]. Coacervation refers to the phase separation in a macromolecu-
drug delivery technology to improve the side effects of orally adminis- lar system in which two phases are formed. The core ingredient is firstly
tered drugs. Reduced irritation system has been designed to reduce suspended or dispersed in a solution of wall material in prior to any
the gastrointestinal irritation induced by certain pharmaceuticals in- droplet formation steps. Coacervation starts by an addition of acid or
cluding aspirin and many antibiotics. This system is composed of an salt (pH change), an input of non-solvent to the dispersion, or a temper-
outer gelatin capsule for quick decomposition in stomach to deliver ature change of the emulsion, resulting in a precipitation of wall
multiple small spherical polymeric pellets containing the active com- material and a continuous coating of wall polymer around the core
pound [67]. The polymeric controlled release system was designed droplets. The wall coating of formed microcapsules can be hardened, so-
to deliver the temozolomide to treat the brain cancer glioblastoma lidified and stabilized by a polymer crosslinking process through a
multiforme in order to reduce the adverse effect of temozolomide. change of temperature or an input of crosslinker. Generally, simple co-
Temozolomide kills the tumors through the methylation of tumoral acervation needs a single colloidal solute while complex coacervation
DNA. Methylation leads to DNA damage and activates apoptotic implicates more than one colloid [16,23,78,85,86]. Ionotropic gelation
pathways in the cell cycle, thus resulting in cell death. The potential depends on the ability of polyelectrolytes to crosslink in the existence
cytotoxic effect could be generated during the antitumor process. In of counter ions to produce the spherical crosslinked hydrogel beads.
vivo animal results revealed that localized intracranial delivery of temozo- The hydrophilic beads are generated by an addition of drug loaded an-
lomide from microcapsules could permit much higher effective drug ionic polymeric drops into an aqueous solution of polyvalent cations.
doses and reduce the systemic toxicity, the survival rate of animals The diffusion of cations into the polymeric drops leads to a three-
could be eventually extended [75]. Microencapsulation also improves dimensional lattice of ionically crosslinked moiety. The mechanical
the handling and the workability of drugs during processing as liquids strength and permeability of the beads can be enhanced by an input
can be prepared into powder or solid forms. This makes pharmaceutical of polycations or polyelectrolytes to the bead surface [86–88].
Table 1
Microencapsulation methods for oral and transdermal drug deliveries.

Category Microencapsulation Particle Stability Advantages Disadvantages Examples


method size (μm)

Chemical Interfacial 0.5–1000 Based on the types of polymeric • High drug encapsulation efficiency [93] • Difficult to control polymerization • Dimethyl-dodecyl-(5-hydroxy-pentyl)-ammonium bro-
polymerization [91] wall materials used [92] reactions, loss of water soluble drugs mide (antibacterial agent) grafted polyurea microcapsules for
caused by exhaustive washing steps topical delivery [94]
Free radical – [92]; • Insulin (core drug, peptide) loaded poly(methacrylic acid)/
polymerization • More side effects after drug poly(ethylene glycol) microparticles for oral delivery
administration due to an involvement [80–82,95]
of nonbiodegradable wall materials or

P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45


residual solvents [19]
Physico- Coacervation 2–1200 An addition of non-solvent forms • Simple and cheap method for drug encapsulation[16]; • Aggregation of microspheres and • Insulin (core drug, peptide) loaded mucin/sodium alginate
chemical (phase separation) [91] stable coacervates without • High drug encapsulation efficiency and efficient hard scaling-up for mass production (wall) microcapsules for oral delivery [98]
causing agglomeration [96] control of the particle size with a narrower size [97] • Hydrocortisone succinate acid (core drug, steroid
distribution [92] hormone)/5-fluorouracil (core drug, anticancer agent)/
phyllanthin (core drug, plant bioactive) loaded chitosan
(wall) microcapsules for oral and topical deliveries [12,14]
• Fucoxanthin (core drug, xanthophyll) loaded fish gelatin–
gum arabic (wall) for oral delivery [99]
Ionotropic gelation – Low mechanical strength due to • No requirement of organic solvents or elevated • Relatively fast release of drug due to • Curcumin-phytosome (core drug, plant bioactive) loaded
permeable hydrogel membrane temperatures; the low mechanical strength [92] chitosan (wall) microspheres for oral delivery [100]
of microspheres [92] • Prevention of liable drug damages such as proteins; • Egg yolk immunoglobulin (core drug, antibody) loaded
• Simple, fast and cost-effective method [92] chitosan/alginate (wall) microcapsules for oral delivery [101]
• Insulin (core drug, peptide) loaded chitosan/alginate
microcapsules for oral delivery [102,103]
Mechanical Fluid bed coating 20–1500 Based on the thickness of the • Total control over the drying temperature; • Relatively longer duration (up to 2 h) • Berberine HCl (core drug) loaded hydroxypropyl–
[91] coating layers [78,91] • Lower operation cost [104] involved in the drying process [104]; methylcellulose microparticles coated with Eudragit E100
• Agglomeration/sticking of particles (wall) for oral delivery [68]
during coating [105] • Diclofenac sodium (core drug, anti-inflammatory agent)
loaded Eudragit RS30D (wall) microcapsules for oral delivery
[106]
Solvent 0.5–1000 Microcapsules prepared with • Useful method for the delivery of small drug • Low drug encapsulation efficiency • Cidofovir (core drug, antiviral agent) loaded poly(lactide-co-
evaporation/ [91] solvent extraction result in higher molecules [92] and high cost [92]; glycolide) (PLGA) microcapsules for topical delivery [107]
extraction porosities than those produced by • More side effects after drug • Human recombinant insulin (core drug, peptide) loaded
solvent evaporation [85] administration due to an involvement poly(D,L-lactide-co-glycolide) (PLGA) (wall) microcapsules
of non biodegradable wall materials or for topical delivery [108]
residual solvents [94] • Carvedilol (core drug, antihypertensive agent) loaded
Ordered mesoporous carbon (wall) microparticles for oral
delivery [109]
Spray drying 5–5000 High stability [110] • Low operation cost, good stability and retention, and • Particle agglomerations and larger • Cidofovir (core drug, antiviral agent) loaded poly(lactide-
[91] easy scaling-up for mass production [110,111]; sized microcapsules [91]; co-glycolide) (PLGA) microcapsules for topical delivery [107]
• No limitation of polymer choices, high encapsulation • Loss of input materials, change of • Ibuprofen (core drug, anti-inflammatory agent) loaded
efficiency, time-efficiency, useful for heat-sensitive drug polymorphism of spray-dried drugs, gelatin (wall) microcapsules for oral delivery [113]
entrapment such as proteins or peptides due to an and limited amount of polymer and • Soft mannitol/lecithin agglomerates containing
involvement of mild temperatures [112] drugs being encapsulated [92]; pantoprazole (core drug, proton-pump inhibitor) loaded
• No uniform microcapsules [110] Eudragit S100 (wall) microparticles for oral delivery [114]

29
30 P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45

4.3. Mechanical microencapsulation processes patients treated with SMANCS had tumor shrinkage (95%) and de-
creased α-fetoprotein levels (86%) [124–126]. G4-polyamidoamine
Fluid bed coating refers to a process that solid drug particles are (PAMAM) dendrimer–ibuprofen conjugates were produced through
suspended on a jet of air followed by spraying a liquid coating on the ester, amide, and peptide linkages. Amide- and ester-linked conju-
drug particles, and the coated wall is solidified through solvent evapora- gates were shown to be relatively stable under the cathepsin B buffer
tion or cooling procedures [78,85,86]. Wurster in 1953 developed the and diluted human plasma release mediums [127]. A novel paclitaxel
coating technique by using a coating chamber with a cylindrical nozzle conjugate was synthesized by using valine–citrulline dipeptide (Val–
and a perforated bottomplate for spraying the coating material on the Cit), a substrate of Cathepsin B (C B ), and p-aminobenzylcarbonyl
core particles [89]. Solvent evaporation/extraction forms the microcap- (PABC) as a spacer, to link polyethylene glycol (PEG) and paclitaxel
sules by dissolving or dispersing the core drug in the coating polymer for the enhanced antitumor activity [128]. Protein stabilization
solution, followed by the dispersion of core-wall solution in a liquid could be achieved by a suitable polymer for microencapsulation sys-
vehicle with agitation [78,86]. The coating material then shrinks around tem to reduce the protein aggregation [129,130] and minimize the
the core drug to produce the microcapsules by removal of the solvent water content of proteins [131]. Protein inactivation and aggregation
from the polymer droplets either by solvent evaporation (by heat or could occur easily during the harsh encapsulation procedures [132].
reduced pressure), or by solvent extraction (with a third liquid which A stabilization method was developed for the encapsulation of γ-
is a precipitant for the polymer and is miscible with both water and chymotrypsin and HRP in Poly( D ,L -lactic-co-glycolic) acid (PLGA)
solvent) [85]. Spray-drying method was industrially employed since microspheres with the use of PEG as a protein stabilizer. Co-
1927 [90]. The core particles are firstly dispersed in a wall polymer solu- lyophilizing of γ-chymotrypsin with PEG before PLGA encapsulation
tion and then sprayed into a hot chamber. The wall material solidifies resulted in a significant decrease in the formation of buffer-insoluble
onto the core particles because the input solvent evaporates and there- aggregates during microencapsulation process, with only 8% protein
fore the microcapsules can be formed in a polynuclear or matrix type. aggregation, as compared with lyophilizing of γ-chymotrypsin in
Spray-drying is normally used for encapsulating labile drugs due to its PLGA microspheres, with more than 30% aggregation and loss of
short contact time in the drier. Spraying drying can be applied with half activity [133]. Glycol chitosan (GC) was incorporated into
the use of supercritical carbon dioxide for the entrapment of sensitive PLGA microparticles in order to stabilize lysozyme as a model pro-
drugs such as proteins [16,23,78,85]. tein. The viscous GC in an aqueous phase for lysozyme entrapment
suppressed the protein hydration and declined the protein leakage
5. Stabilization of drugs using microencapsulation technology during microcapsule formation process. Microcapsules with a higher
amount of GC remained the bioactivity of protein, with more than
Drug stability refers the capability of a drug formulation in a spe- 95 wt.% of lysozyme remaining for 28 days, whereas the control mi-
cific container/closure system to preserve its own properties and crocapsules revealed a great decrease in protein bioactivity, with
therapeutic effects throughout the period of storage and usage about 92 wt.% after only 7 days [134].
[115]. In fact, many drugs are liable to chemical and physical degra- Many antibiotics become unstable under gastrointestinal environ-
dation due to the fragility of their molecular structure. Chemical deg- ment after oral administration, resulting in low bioavailability. The
radation (pH, moisture, oxidation) and physical degradation antibiotic stabilization could be accomplished by the combination of a
(temperature, light) of drugs may alter their pharmacological func- non-toxic polymeric material and the antibiotic to stabilize the antibiotic
tions, leading to a changed therapeutic efficiency and toxicological under aqueous conditions. Crosslinked polymeric complex could give
consequences [116]. For examples, β-lactam antibiotics, especially a protective coating against the harsh environment of the stomach and
penicillin, easily degrade in the presence of acidic aqueous condi- increase the chemical stability of acid-degradable moieties/drugs [135].
tion [117]. Vitamin C is susceptible to oxidation [118]. Protein- A gastro-mucoadhesive chitosan/alginate system was designed to stabi-
based drugs are highly unstable to pH and temperature variations, lize and protect the antibiotic amoxicillin under the gastric condition
moisture and oxidation [119]. Nifedipine, epirubicin and idarubicin after oral administration. Microencapsulated amoxicillin was demon-
are sensitive to light [120,121]. The drugs should be stable to remain strated to be highly stable under the simulated gastric fluid, with the
their quality until the time of usage. The drug quality could be kept drug degradation of only 7.6% after 8 h, when compared with the plain
by using the microencapsulation technology which provides a qual- amoxicillin, with 90.9% drug degradation [136]. Clarithromycin loaded
ity protection of drugs using the polymeric micro-carriers. Microen- Eudragit L100 microcapsules were developed in order to increase the
capsulation offers a quality maintenance and preservation of drug stability and avoid the fast dissolution of drug after oral administra-
susceptible drug molecules to stabilize the sensitive drugs from the tion. Eudragit L100 microcapsules did not show significant changes in
external environment. Pharmaceuticals are conjugated or embedded the drug content of acidic suspension of optimal batch. The prepared for-
in the polymeric wall materials of microcapsules or microparticles mulation revealed a good stability for 14 days [137]. The drugs used for
for the drug stabilization and the controlled drug release [122]. Ther- the treatment of Acquired Immunodeficiency Syndrome (AIDS) includ-
apeutic agents could be prevented from interacting with the external ing lamivudine and stavudine are unstable with short half-life after oral
molecules which may deteriorate the nature of drugs and lead to an administration. Both drugs became stable at 40 °C with 75% RH for
inefficiency of their bioactivity. Crosslinked polymer chains in the about 3 months with less drug loss when they were encapsulated within
wall matrix or wall shell of microcapsules could delay the drug disso- the cellulose-based microcapsules [138,139]. Microencapsulation tech-
lution under gastrointestinal fluids by forming the diffusion barrier nology has also been applied to other therapeutic compounds to enhance
to stabilize and protect the inner drugs. The polymeric wall of micro- their half-life and stability. Ubiquinone-10 (CoQ10), a vitamin-like lipo-
capsules is subject to the initial hydrolysis and enzymatic cleavage philic compound used in the pharmaceutical industries, is vulnerable
which lead to the scission of polymer chains [123]. Drugs could be to light, oxygen and temperature. CoQ10 microcapsules prepared with
covalently bound to the polymer chain or backbone to produce a 20% gum arabic as the wall material resulted in a high increase in the
drug-polymer conjugate for a prolonged delivery of unstable drugs drug half-life of 693 min when compared with the free CoQ10, with
such as proteins. The first polymer–protein conjugate SMANCS the half-life of 111.17 min for at 30 ± 2 °C under UV light [140].
(Zinostatin stimalamer®) was developed in 1990 and available com- Nattokinase (NK), an effective fibrinolytic enzyme, was entrapped in
mercially. Maeda and Konno synthesized the conjugate which high molecular weight γ-polyglutamic acid in sodium form (Na-γ-
consisted of an anti-tumor protein neocarzinostatin (NCS) covalent- PGA) microcapsules in order to improve the temperature and pH stabil-
ly bound to two polymer chains of styrene-co-maleic anhydride ity. NK became more stable over a broad range of pH (from 2.0 to 12.0),
(SMA). The clinical study showed that a majority of hepatoma at higher temperatures (50 °C and 60 °C) and under acidic conditions
Table 2
Summary of polymeric material-based microencapsulation systems for the applications of oral and transdermal drug deliveries.

Polymeric materials Delivery Entrapped drugs Biomedical applications References


route

Chitosan Oral Curcumin Curcumin-phytosome loaded chitosan microspheres had a prolonged half life and improved oral absorption of curcumin when compared with those of curcumin [100]
phytosome and curcumin loaded chitosan microspheres.
Chitosan Oral Celecoxib Microcapsules had a better inhibition of cyclooxygenase-2 protein expression from mice hepatocytes than free celecoxib. [160]
Chitosan Oral Hydrocortisone succinic ACTH concentration in microcapsule mice plasma was more than 40% lower than that of water control. [12]
acid
Chitosan–alginate Oral IgY Microspheres effectively protected the IgY antibodies against ETEC-induced diarrhea in older pigs. [101]
Chitosan Oral Insulin Phytic acid-crosslinked chitosan capsules improved the intestinal absorption of insulin and remarkably reduced blood glucose levels in diabetic mice when [161]
compared with tripolyphosphate-crosslinked chitosan capsules.
Chitosan–alginate Oral Insulin Blood glucose concentration in diabetic rats could be significantly reduced and stably maintained for a prolonged period after administration of insulin loaded [102]
microspheres when compared with the free insulin.
Chitosan Transdermal Miconazole nitrate/ Drug release raised with an increase in pressure from 0.028 to 5 kg. [162]
clotrimazole
Chitosan Transdermal 5-FU Microencapsulated 5-FU had a stronger growth inhibition of human keratinocytes than free 5-FU. 5-FU could be released from microcapsule treated textiles on an [12]

P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45


intact mice skin.
Chitosan Transdermal Vitamin E Microencapsulated vitamin E significantly increased the skin moisture and elasticity, and also reduced the skin wrinkle and roughness. [163]
Gelatin Oral Ibuprofen Microcapsules caused a significantly higher initial plasma concentration in rats, Cmax and AUC of ibuprofen than the free ibuprofen. [113]
Gelatin–agar Oral Gallic acid Microcapsules were more effective in an oral treatment of acute liver and kidney toxicity in mice than the free gallic acid after an overdose of acetaminophen. [11]
Gelatin Transdermal Vitamin A palmitate Vitamin A palmitate was released at a much slower rate from microspheres containing gel than the conventional plain vitamin A palmitate gel in rat skin. [187]
Gelatin–agar Transdermal Berberine Microcapsules had a better anti-S. aureus activity than the free drug. Berberine could be released from microcapsule treated textiles on nude mice skin. [11]
Gelatin–agar Transdermal Phyllanthus urinaria Microcapsules improved the anti-A. niger activity when compared with the free herbal extract. [188]
Alginate Oral Insulin After 5 h, blood glucose lowering effect caused by micro-particles in diabetic rabbits was equal to that induced by the subcutaneously administered insulin [99]
solution.
Alginate Oral Isoniazid Microspheres could be detected in the intestine of adult Wister rats after 24 h. [199]
Alginate–chitosan Oral Metronidazole Microcapsules had a better eradication of H. pylori infection in mice than the suspension form of free MZ. [200]
Alginate–sweet potato Oral Glipizide Microcapsules possessed a slow, complete and extended release of glipizide, as well as a good mucoadhesive property in the in vitro wash-off test. [201]
starches
Alginate–sodium Oral Gliclazide Microencapsulated drug had a prolonged hypoglycemic effect (2–16 h) in diabetic albino rats when compared with the free drug (0.5–10 h). [203]
carboxymethyl cellulose
Alginate–methyl cellulose Oral Gliclazide Gliclazide-loaded microcapsule-treated alloxan-induced diabetic albino rat group showed a significant hypoglycemic effect when compared with the free drug. [204]
Ethylcellulose Oral Salbutamol sulfate Oral bioavailability of microcapsules was demonstrated by young healthy human volunteers. [205]
Ethylcellulose Oral Clarithromycin Microcapsules did not decrease the bioavailability and had a better therapeutic performance with a prolonged drug release in healthy volunteers. [206]
Trehalose/hydroxyethyl Transdermal Vancomycin Microencapsulated vancomycin could be useful for topical applications on the potential microbial contamination on skin wounds. [207]
cellulose
PLA Oral Insulin Insulin loaded PLA microcapsules were capable of lowering the blood glucose level in the diabetic rat. [214]
PLGA Oral Amifostine Half of amifostine from PLGA microcapsules was released after the first 6 h, and 92% of amifostine was further released after 12 h. [215]
PLGA Oral Plasmid DNA Microcapsules loaded with plasmid DNA could induce the immune system to produce the corresponding fish-anti-LCDV antibody which could be detected in [216]
the sera of Japanese flounders.
PLGA Transdermal Acyclovir Acyclovir reservoir in the porcine skin basal epidermis treated with microparticles was higher than that of control suspension after 88 h, suggesting that PLGA [217]
microparticles could improve acyclovir topical therapy as the drug retention in the basal epidermis.
PLGA Transdermal Cidofovir Higher amount of cidofovir was found in the basal epidermis treated with microparticles than that of control solution, revealing that cidofovir-loaded micro- [218]
particles could increase the drug retention in the basal epidermis.
PLGA Transdermal Human recombinant Released insulin from PLGA microspheres stimulated a rapid cell migration following an induced scratch as fresh insulin over 23 days, suggesting a high level of [108]
crystalline insulin sustained bioactivity.
Locust bean gum-PVA Oral Buflomedil HCl Microspheres could offer a controlled release of buflomedil HCl with short half life and high water solubility. [221]
Lepidium sativum-PVA Oral Simvastatin Microspheres could provide a controlled release of simvastatin from the crosslinked polymer matrix. [222]
Gum ghatti-PVA Oral Ranitidine HCl Microsphere prepared with higher amount of glutaraldehyde and PVA resulted in a higher mucoadhesivity and a slower drug release. [223]
PCL Oral Bovine serum albumin Bovine serum albumin protein could keep unchanged with no aggregates during the encapsulation process. [227]
PCL Oral Manidipine Drug loaded PCL microparticles could reduce the mean arterial pressure variation up to 24 h after phenylephrine injections when compared with the pure drug [228]
dihydrochloride and drug loaded poly(3-hydroxybutyrate-co-3-22 hydroxyvalerate) microparticles.
Eudragit® L100-55 Oral Anti-VP8 IgY Microcapsules had the retention of more than 95% of IgY activity after 6 h incubation in SGF when compared with the free IgY, with a significant decrease (86%) [233]
in IgY activity after 30 min. In vivo weaned pig model revealed microencapsulated IgY exhibited a much higher stability after oral administration.
Eudragit® L30 D-55 Oral Vibrio cholerae C7258 Oral administration of 3.5 mg microencapsulated vibrio cholerae C7258 inactivated strain showed an appropriate protection of the bacteria which induced the [234]
inactivated strain production of high levels of vibriocidal titers in adult rats as compared to the same amount of non-encapsulated one.

31
32 P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45

(pH 5 and 1.2) as opposed to the free NK. Microencapsulated NK also had Kim and Park [147] reported that the tulobuterol release from PCL/
more protective bioactivity than the free NK [141]. Eudragit RS 100 microcapsules was faster than that of PCL microcapsules
in the succinic acid release medium because the hydration level increased
6. Drug release mechanisms in the presence of Eudragit RS 100. The increased hydration level of mi-
crocapsules might be associated with the fact that the quaternary ammo-
6.1. Diffusion nium groups of Eudragit RS 100 interacted with the carboxyl group of the
succinic acid. With the higher hydration level, the swelling and dissolu-
Drug release can be done by drug diffusion from polymeric reservoir tion of microcapsules increased. This increased the drug diffusion from
microcapsules or polymeric matrix microcapsules. In the reservoir sys- the microcapsules through an increased permeability of the microcapsule
tem, the drug is entrapped within the core reservoir by a polymeric wall. Kulkarni et al. [148] investigated the relationship between the re-
wall shell. The drug first partitions into the polymeric wall of microcap- lease of diltiazem HCl and the crosslinking extent of gellan gum/egg albu-
sules from the core reservoir, and then diffuses out to other side of the min microcapsules. Diltiazem HCl was first bound to a cation exchange
wall shell in the medium. When the core reservoir is saturated and a resin Indion 254® before microencapsulation in order to reduce bitter
constant concentration gradient of the drug is remained in the wall taste of drug and increase the stability by protecting the drug from hydro-
shell, the rate of drug flux is constant. This results in zero order drug re- lysis. The drug-resin complex showed an extended dissolution up to 3 h
lease. The drug release rate maintains constant as long as the internal when compared with the free drug (1 h). The crosslinked microcapsules
and external concentrations of core drug and the concentration gradient could prolong the drug release up to 15 h. A decrease in microcapsule
through the wall shell are constant. Finally, the drug concentrations in swelling was promoted by a higher amount of glutaraldehyde which
the core reservoir and in the wall shell fall below saturation, the release may produce a stiffer polymer network during microcapsule formation
rate of drug declines [23,142]. Kooiman et al. [143] reported the devel- process. This reduced the water adsorption and the volume of the poly-
opment of oil-filled poly(L-lactic acid) (PLA) terminated with 1H–1H meric matrix. The drug transport through the polymer was then retarded.
perfluoro-octan-1-ol microcapsules for the ultrasound-triggered deliv-
ery of lipophilic drugs. Hexadecane oil was used as the core drug carrier 6.3. Erodible and degradation
reservoir with cyclodecane gas, while the absorbing dye Sudan Black, as
a hydrophobic molecule, was selected to mimic a lipophilic model drug. Erosion and degradation of a drug delivery system should involve the
Sudan Black release was efficient for half oil-filled microcapsules since nontoxic, biodegradable and biocompatible polymeric materials. The
75 ± 6% of the amount encapsulated was released with the oil. The drug is homogeneously mixed with the polymer in the microencapsula-
remaining Sudan Black was retrieved from the bottom fraction, suggest- tion system. Erosion and degradation of the polymeric wall of microcap-
ing that it stayed with the shell. In the matrix system, the drug is firstly sules highly decrease the distance between the drug molecules and the
encapsulated in the polymeric matrix of microcapsules and the drug re- microcapsule surface. This results in the drug release without transport.
lease is achieved by diffusion through the matrix. This system involves However, the hydration process is usually quicker than the erosion and
the initial burst release of drug from the wall matrix surface. The drug degradation of polymeric matrix of microcapsules, causing diffusion
release rate depends on the physical properties of polymer used for and release of the entrapped drug through micro-pores of microcapsules
the matrix wall. Generally, the hydrophobic polymers delay the release induced by water uptake. The polymeric matrix begins to be eroded and
and diffusion of the drug from microcapsules [23,142]. Jeong et al. [144] degraded but the encapsulated drug molecules still maintain in the inte-
investigated the drug release of papaverine loaded PCL microparticles. rior of microcapsules. Eventually, the entrapped drug is released once the
There was a small initial burst release of papaverine from microcapsules polymeric matrix disintegrates [23,142,149]. Zhu et al. prepared the
due to the drug encapsulated near the surface of hydrophobic PCL wall. felodipine-loaded PLA/polymerized-N-maleoylchitosan (p-NMCS) core–
The subsequent drug diffusion through the amorphous region of PCL shell microcapsules and the drug release mechanism was studied. The re-
wall matrix into the release medium was delayed. The characteristics sults showed that the drug release of p-NMCS microcapsules was driven
of PCL also had an influence on the drug release rate. Lower PCL concen- by a diffusion process, while the drug release of PLA/p-NMCS microcap-
tration (2.5 wt.%), higher molecular weight (80 K) of PCL and less per- sules with high ratio of PLA to p-NMCS (not less than 1/1) was propelled
fectly shaped crystalline lamellae of PCL wall matrix also contributed by a combined diffusion and degradation process. The encapsulated
to a faster rate of drug release. felodipine was slowly released from the PLA/p-NMCS core–shell micro-
capsules as a result of PLA degradation [23,150]. Erosion or degradation
6.2. Swelling and dissolution of the polymeric matrix could be induced by pH or enzymatic hydrolysis
which leads to the release of entrapped drug [151]. The release of 5-
Swelling involves the water uptake by a polymer system and the poly- Aminosalicylic acid from the crosslinked chitosan–sodium cellulose sul-
mer system then increases in its volume. Swelling of a polymer is closely fate–sodium polyphosphate microcapsules in the simulated colonic
related to the drug dissolution from the polymer system. When swelling fluid (pH 6.4) with glucanglucanohydrolase and phosphate buffer
happens in the drug loaded polymeric matrix, the water enters the poly- (pH 6.4) was discussed [152]. The micro-pores could be found in the
meric matrix and the polymer system swells by the uptake of water. At surface of microcapsule film which was subject to the simulated co-
this stage, the drug dissolution occurs, the entrapped drug gradually dis- lonic fluid (pH 6.4) with glucanglucanohydrolase after 12 h because
solves and then diffuses through the swollen polymeric matrix to the re- of enzymatic erosion or degradation of microcapsule film. The swell-
lease medium [23,142]. The swelling rate of the polymer system can be ing of microcapsules was reduced and the erosion or degradation
governed by the properties of polymeric materials. Generally, the drug- was increased due to the presence of enzyme in the release medium.
polymer formulation contains a hydrophilic polymer with low density With an increased erosion or degradation, the drug holding capacity
of crosslinking chains, water penetration leads to a rapid swelling of the of microcapsules was decreased. This resulted in more amount of
polymer, resulting in the faster drug dissolution from its present solid drug release after 12 h when compared with the microcapsules in
phase to the release medium [142,145]. Fundueanu et al. [146] synthe- phosphate buffer (pH 6.4) medium.
sized the cellulose acetate butyrate (CAB) microcapsules containing
sulfopropylated dextran microspheres loaded with tetracycline HCl. The 6.4. Osmosis
drug release was governed by the wall shell rupture of CAB microcapsules
under the pressure caused by the swelling of the encapsulated dextran Drug release related to osmosis refers to the movement of water
ion exchange resins. The force induced by swelling then broke the more molecules down the concentration gradient through the semiperme-
resistant wall of microcapsules, resulting in self-propelled drug release. able wall shell of microcapsules until there is a balance between both
P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45 33

sides. The rate of drug (osmolyte) flowing across the wall shell of micro- transportation [158]. Chitosan is considered as a safe material for
capsules depends on the drug concentration and property, the perme- microcapsule-based oral drug delivery carrier that protects the liable
ability of wall shell and temperature. Release medium passes through drug from the gastrointestinal environment in a number of previous
the microcapsule wall and dissolves the entrapped drug, causing a con- studies. Zhang et al. [100] developed the curcumin-phytosome loaded
centration gradient which attracts more water inside [23,142]. Osmotic chitosan microspheres for oral administration. The pharmacokinetics
pumping happens when the osmotic pressure, induced by water ab- study showed that the half-life of curcumin-phytosome loaded chitosan
sorption, propels the transport of the drug [149,153]. In the osmotically microspheres (3.16 h) was longer thanboth curcumin-phytosome
related drug release mechanism, the osmotic flux of water passes across (1.73 h) and curcumin loaded chitosan microspheres (2.34 h). The re-
the wall and dilutes the drug solution inside the microcapsules. This re- sults revealed that curcumin-phytosome loaded chitosan microspheres
sults in swelling, microcracks, and eventually the rupture of microcap- exhibited a better sustained drug release and an improved oral absorp-
sules. When cracks are formed, drug solution leaks through them and tion of curcumin than others. The study suggested that a carrier combin-
interconnects microcapsules that have been already ruptured [154]. A ing chitosan microspheres and phytosomes, could be a more promising
biodegradable micro-osmotic pump was developed for the controlled vehicle for oral administration of curcumin. Cheng et al. [160] reported
release of basic fibroblast growth factor (bFGF). The bottom layer of the chitosan-based microcapsules containing celecoxib as a cyclooxy-
the device contained a reservoir for drug storage and micro-channels genase 2 inhibitor. The possible therapeutic activity of celecoxib con-
for drug release. Another layer of the 85/15PLGA was considered as a taining microcapsules on mice was investigated after oral
semi-permeable layer. When the device was immersed in an aqueous administration by examining the immunohistochemistry section of
condition, osmotic pressure drove water into the reservoir and the the liver. The celecoxib containing chitosan microcapsules showed a
internally-built pressure started to drive the drug (bFGF) through the better inhibition of cyclooxygenase-2 protein expression from mice he-
micro-channels [155]. Another biodegradable device was constructed patocytes when compared with an oral administration of free celecoxib.
for the controlled release of ciprofloxacin-HCl (CIP). This device Plasma liver functional enzyme markers and histochemistry analysis re-
consisted of osmosis and diffusion mechanisms to release CIP for vealed that chitosan-based micro-particulate system did not have toxi-
short-term therapies. The poly(glycerol-co-sebacic acid) (PGS) was lo- cological adverse effects on the mice hepatocytes. Lam et al. [12]
cated in a tubular geometry with solid drug powder packed into its developed the chitosan-based microcapsules to deliver hydrocortisone
core and a micro-orifice was drilled through its wall in order to expulse succinic acid through oral administration. The potential of chitosan-
the drug from the device. PGS acted as a semipermeable material for the microencapsulated hydrocortisone succinic acid for oral application
elementary osmotic pump to accomplish the release of CIP [153]. was demonstrated using ACTH assay from C57BL/6 mice. The mean
ACTH concentration in drug loaded chitosan microcapsule mice plasma
7. Recent advances in microencapsulated oral and transdermal drug was detected to be approximately 42.2% lower than that of water con-
delivery systems trol. Li et al. [101] reported the use of chitosan–alginate microcapsules
to protect the egg yolk immunoglobulin (IgY) against K88 + ETEC
In fact, microcapsule-based drug delivery systems offer many (enterotoxigenic E. coli)-induced diarrhea using an in vivo pig model.
benefits to drug delivery, targeting and release. Their great potential Chitosan–alginate microspheres could effectively protect the IgY anti-
to integrate with conventional drug dosages leads to the emergence bodies from gastric inactivation after oral administration, resulting in
of effective micro-medicines for oral and topical treatments. The use an enhanced protection against ETEC-induced diarrhea in older pigs.
of natural and synthetic polymers as drug carriers in the microencap- Since the protein- and peptide-based drugs were highly susceptible to
sulation systems is possible to improve and enhance the drug stabil- the gastrointestinal environments, some efforts were put on this area.
ity in biological environments including the gastrointestinal tract Lee et al. [161] compared the therapeutic performance between phytic
and the first pass metabolism after oral administration, as well as acid- and tripolyphosphate-crosslinked chitosan capsules for insulin de-
skin layers during transdermal administration. This also increases livery. Phytic acid-crosslinked chitosan capsules resulted in an im-
the drug entrapment efficiency, and utilizes drug targeting, release, proved intestinal absorption of insulin and significantly decreased
absorption and interaction with targeted body sites. Recently, some blood glucose levels in diabetic mice when compared with the
novel microencapsulation systems have been developed in order to tripolyphosphate-crosslinked chitosan capsules. Zhang et al. [102] ex-
provide more choices for drug entrapment. Table 2 summarizes the amined the chitosan–alginate microcapsules for the oral delivery of in-
polymeric material-based microencapsulation systems for oral and sulin. The blood glucose concentration in diabetic rats could be
transdermal drug deliveries evaluated by in vivo and in vitro models. significantly reduced and stably remained for 96 h after oral adminis-
tration of insulin loaded microspheres when compared with the free in-
7.1. Natural polymer-based systems for drug microencapsulation sulin (Fig. 2). The results suggested that the alginate–chitosan
microspheres could be a potential carrier of protein or peptide drugs
7.1.1. Chitosan-based systems in oral administration.
Chitosan is a cationic linear polysaccharide composed of β(1–4) Transdermal delivery of drugs using chitosan microcapsules as car-
linked glucosamine with N-acetylgucosamine. It is originated from ex- riers has also been investigated. Yuen et al. [162] reported the potential
tensive deacetylation of chitin existed in shells of crustaceans. There use of chitosan-based microcapsules for the topical delivery of two
are different obtaining procedures that can affect the soluble properties antifungal agents, miconazole nitrate and clotrimazole, for the tinea
of chitosan. Chitosan must be dissolved in the media with acidic pH, and pedis treatment. The release rates of two drugs were investigated
using 1 to3% aqueous acetic acid solution to solubilize chitosan is very under different pressures from 0.028 to 5 kg at different pH values
common. This is because chitosan contains the amino group which is from 5.5 to 8.5. The drug release increased with an increase in pressure
water-insoluble, positively charged, and only soluble at the state of and the release of drugs had no significant difference at different pH
pH b 6. Chitosan processes many advantages in microcapsule forma- values. Lam et al. [12] demonstrated the topical delivery of 5-FU using
tion. Chitosan is a natural polymer which is biodegradable, chitosan-based microcapsules. Chitosan-based microcapsules contain-
biocompatible, non-toxic and safe. It is able to control the release profile ing 100 μg/mL of 5-FU resulted in a stronger growth inhibition of
of active ingredients [10,12–15,90,156,157]. Chitosan is a mucoadhesive human keratinocytes when compared with the free drug. The skin
polymer that increases residence time at the site of drug adsorption and cells with both untreated control (Fig. 3a) and blank microcapsules
the half time of drug clearance, and thereby improving the drug without drugs (Fig. 3b) showed a high integrity of cellular morphology
bioavailability [158,159]. A higher degree of deacetylation (N 65%) can while cells with both 5-FU microcapsules (Fig. 3c) and free 5-FU
increase the charge density, and eventually improve the drug (Fig. 3d) had certain degree of cellular damage. The 5-FU microcapsules
34 P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45

could be released from chitosan-based microcapsule treated textiles


on the intact mice skin model. Yenilmez et al. [163] prepared the vita-
min E containing chitosan microspheres for topical delivery. In vivo
study conducted on human cheeks and crows' feet skin demonstrated
that the vitamin E microspheres significantly increased the skin mois-
ture and elasticity, and reduced the skin wrinkle and roughness.
Chitosan is regarded as a promising candidate for oral drug delivery
due to its mucoadhesive property. Chitosan-based micro-particulate
system containing drug loaded oil is believed to be useful in oral drug
delivery. Most conventional drugs have poor bioavailability and absorp-
tion when they are orally administered because of drug degradation
under the gastrointestinal environment and the first-pass metabolism.
Therefore, many drugs are required to be taken frequently in high
doses in order to achieve an effective therapeutic level. Frequent
drug administration in high doses can induce toxicity and other side
effects [164]. As a result, the optimal drug formulation prepared with
mucoadhesive chitosan might minimize the in vivo drug degradation,
Fig. 2. The serum glucose concentration after oral administration of insulin-free and
offer a drug release over an extended period of time and also reduce
insulin-loaded (100 IU/kg) alginate–chitosan microspheres to STZ-induced diabetic rats.
Statistically significant difference from insulin-free microspheres: *p b 0.05; **p b 0.01. the potential toxicity of drugs. Mucosal membranes are the moist sur-
(Adapted with permission from [102]). faces of wall linings existing in various body cavities including eyes,
nostrils, buccal cavity, respiratory, gastrointestinal and reproductive
tracts. These membranes offer the lubrication and control the moisture
exhibited a stronger cell inhibitory effect than the free 5-FU (Fig. 3e). content of the cell epithelial surfaces [165–167]. Mucoadhesion is con-
This might be due to the sustained release of 5-FU from microcapsules, sidered as an attractive interaction at the interface between a drug
resulting in a continuous therapeutic effect against the growth of skin and a mucosal membrane [166]. Mucoadhesive property of a pharma-
keratinocytes. In vitro drug delivery model also showed that 5-FU ceutical formulation can be accomplished by introducing hydrophilic
polymers with good sticking ability during the formation process. Chito-
san has both primary amine and hydroxyl functional groups which en-
hance hydrogen bond formations such as \OH and \COOH [168]. The
linear molecule of chitosan displays enough chain flexibility and its sur-
face energy characteristics prefer to spread into the mucosal surface.
The chitosan conformation depends on its ionic strength. All of them
determine the mucoadhesion activities [168–171]. Chitosan has an ex-
cellent mucoadhesion among various polymers and it possesses the
mucoadhesive activity due to the ionic interaction between the posi-
tively charged primary amino groups in chitosan and negatively
charged sialic acid and sulfonic acid substructures of the mucus in cell
membrane [158,172,173]. This interaction extends the residence time
at the site of drug absorption due to the increased contact with the ab-
sorbing mucosa, leading to a steep concentration gradient to favor drug
absorption and localization in specified regions to enhance the drug bio-
availability [174,175]. Previous studies have shown that the presence of
chitosan demonstrated an improved mucoadhesion [167,176–179].
Chitosan has also been reported to enhance the mucosal permeation
which resulted in the improvement on drug adsorption [180]. There-
fore, chitosan is considered as a suitable polymer to produce the
micro-carriers for oral drug delivery to protect the susceptible drugs
such as protein- and peptide-based medicines from the gastrointestinal
environment for increasing drug bioavailability and drug adsorption.
Oral delivery of chemotherapy still remains challenges in pharmaceuti-
cal and therapeutic fields. Chemotherapeutic drugs are generally ad-
ministered through intravenous injection and this results in patient
discomfort and inconvenience. Oral administration of chemotherapy
can significantly damage the esophagus, stomach as well as other or-
gans. The availability of oral chemotherapeutic dosage forms would
greatly improve the life quality and compliance of patients with mini-
mal disruption of activities of their daily living [3,181]. The dual protec-
tion of the chemotherapeutic agent could be provided by an internal oil
phase together with a chitosan-based micro-carrier. It is believed that
the internal oil phase of microcapsules might protect the anti-cancer
drug from the gastrointestinal environment during oral administration.
The chitosan wall shell of microcapsules might be gradually dissolved in
Fig. 3. Growth inhibition towards skin keratinocytes: (a) untreated control; (b) blank mi- the stomach. The bile might emulsify the oil into smaller oil droplets
crocapsules; (c) 5-FU microcapsules; (d) 5-FU; (e) percentage of cell viability. Each exper-
iment was done in triplicate and a mean value was obtained while three independent
which are called “micelles”. These micelles are believed to protect the
experiments were performed. Results are shown as mean ± SD from their mean values chemotherapeutic agent from the gastrointestinal environment and
of these three independent experiments. (Adapted with permission from [12]). thereby avoiding the drug to attack the gut lining (Fig. 4). Since the
P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45 35

Fig. 4. Schematic diagram of the acidic degradation of chitosan-based microcapsules con-


taining drug loaded oil in stomach and the bile emulsification process of drug containing
micelles.

nutrients especially lipids and fats are absorbed in the small intestine
followed by entering the lymphatic circulation, the drug containing
micelles might then enter the lymphatic circulation directly without
the influence of the first-pass metabolism. The drug bioavailability and
drug adsorption would be greatly enhanced by using the oil containing
chitosan-based microcapsules for oral drug delivery. Fig. 5. H and E histochemical analysis of liver and kidney sections from the survival mice
treated with APAP alone (on day 1) followed by single dose daily of deionized water, gallic
acid loaded microcapsules, or gallic acid from day 2 to 4. In water control group, all the
7.1.2. Gelatin-based systems
liver sections exhibited necrosis whereas 2 of 3 the kidney sections exhibited necrosis
Gelatin is a derived protein substance from collagen which is and only mouse 1 showed a certain degree of cellular integrity in its kidney section. It
isolated from the boiled connective tissues, tendons, bones and skins should be also noted that 1 of 3 the liver and kidney sections (mouse 2) from the micro-
of animals, usually fishes, cows and pigs [182]. Gelatin is widely applied capsule treated group exhibited necrosis while those mice treated with gallic acid exhib-
to pharmaceutical areas as it is a natural polymer which is biodegrad- ited necrosis. (Adapted with permission of The Royal Society of Chemistry [11]).

able, biocompatible and non-toxic. Gelatin is able to produce a high


drug loading, and it is easy for administration and removal in physiolog- Gelatin-based micro-particulate systems have also been used in
ical environments [183]. Apart from tryptophan and low in methionine, transdermal drug delivery. Wakode and Baja [187] reported the use of
cystine and tyrosine, all the amino acids exist in gelatin [184]. The amino gelatin microspheres to increase the stability of vitamin A palmitate
acid composition in gelatin varies from one source to another and main- for transdermal delivery. In the rat skin release model, vitamin A palmi-
ly consists of great amounts of glycine, proline and hydroxyproline tate was released at a much slower and linear rate from the gelatin
[185]. Gelatin is a gelling agent that is soluble in hot water and subse- microspheres containing gel when compared with the conventional
quently capable of forming strong, transparentelastic and elastic ther- plain vitamin A palmitate gel. Lam et al. [11] reported the synthesis of
moreversible gels and flexible films on cooling below about 35 °C. a formaldehyde-free gelatin/agar-based microencapsulation system
Generally, there are two types of gelatins, Type A and Type B. Type A gel- for topical delivery of berberine. The Minimum Inhibitory Concentra-
atin is produced by soaking skin or bones in a diluted acidic medium tions (MICs) and antibacterial tests proved that berberine loaded
followed by extraction at acidic pH while Type B gelatin is solubilized gelatin/agar-based microcapsules exhibited a better growth inhibition
at near neutral pH at about 60–90 °C [186]. Gelatin exhibits the desired towards S. aureus when compared with the original drug. The drug de-
properties which make it preferable for drug delivery applications. In livery model (Fig. 6) demonstrated the delivery of berberine from mi-
micro-particulate oral drug delivery, Li et al. [113] formulated a novel crocapsule treated textiles on nude mice skin. Berberine has the
gelatin microcapsules for the encapsulation of ethanol and ibuprofen fluorescent property. If the treated skin site showed the fluorescent ef-
using a spray drying method. The ibuprofen-loaded gelatin microcap- fect after the topical treatment, the berberine could be released from
sules led to a significantly higher initial plasma concentration in rats, the microcapsules, delivered on the skin surface and absorbed by the
Cmax and AUC of ibuprofen than the free ibuprofen powder, revealing skin. As the treated site of nude mice skin showed the fluorescent effect,
that the drug from gelatin microcapsules resulted in more effective ab- this result suggested that the drug loaded gelatin-based microcapsules
sorption after oral administration. Lam et al. [11] designed a could be a useful vehicle for topical drug delivery. Lam et al. [188] also
formaldehyde-free gelatin/agar microcapsules for the oral delivery of studied the anti-A. niger activity of Phyllanthus urinaria (P. urinaria)
gallic acid. The in vivo mice disease model (Fig. 5) revealed that the oral- loaded gelatin/agar-based microcapsules. This novel green gelatin–
ly administered gallic acid loaded gelatin/agar microcapsules were agar microencapsulation system with water extract of P. urinaria was
helpful to alleviate the damages of acute liver and kidney toxicity in shown to exhibit an improved anti-A. niger activity when compared to
mice after an overdose administration of acetaminophen compared the free herbal extract.
with the free gallic acid. The free drug induced the death of 2/3 mice Gelatin/agar-based micro-particulate system could be used for
whereas all microcapsule-treated mice did survive. This might be due transdermal drug delivery. The skin is considered as an ideal biological
to the high dose administration of free gallic acid which might lead to site with a readily accessible and large surface area for drug administra-
the adverse effects and drug toxicity. However, the microencapsulated tion. Transdermal drug delivery can avoid the disadvantages of oral ad-
gallic acid within the gelatin/agar micro-carrier might promote the ministration including enzymatic degradation and rapid clearance in
sustained drug release over a prolonged period of time and this might the gastrointestinal tract and the first-pass metabolism, and overcome
reduced the adverse side effect after a high dose of oral administration. the drawbacks of intravenous injection including pain, discomfort, and
36 P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45

Fig. 6. The nude mice skin experiment was performed to demonstrate the possible delivery
of berberine HCl from microcapsule fabricated cotton: a. nude mouse; b. nude mouse skin
treated with microcapsule fabricated cotton samples; c. nude mouse skin treated with mi-
crocapsule fabricated cotton after 24 h; d. nude mouse skin under UV illumination and e. re-
Fig. 7. Transdermal drug delivery from gelatin-based matrix microcapsules.
lease of berberine HCl from microcapsule fabricated cotton after 24 h. For b, c, and d, the left
sample represents the microcapsule without drugs and the right sample represents the ber-
berine HCl loaded microcapsules. The data in panel (e) represent the average from in vitro
release of berberine HCl loaded microcapsule experiments ± SD from three independent
7.1.3. Alginate-based systems
experiments. (Adapted with permission of The Royal Society of Chemistry [11]).
Alginate, as an anionic linear polysaccharide containing 1,4-linked
D-mannuronic acid and L-guluronic acid residues, is obtained from
inconvenience of patients [142]. However, the skin is a major barrier of brown seaweed using a dilute alkaline extraction. The resulting solution
drug delivery. Therefore, an effective therapy would tackle the chal- is treated with mineral acids and finally converted to sodium alginate
lenges of topical drug delivery to deliver the required amount of drug [195,196]. Sodium alginate is a natural, biocompatible, biodegradable
dosages into the treated skin sites. Gelatin is an amphoteric polymer and hydrophilic polymer. It has been applied to micro-particulate
that simultaneously has cationic and anionic functional groups. Gelatin formulations for drug delivery because of its bioadhesive, gelation and
was reported to exhibit relatively poor mucoadhesive characteristics pH responsive properties [99,197]. Sodium alginate can form hydrogels
like non-ionic polymers due to its amphoteric property and self- with multivalent cations such as Ca2+, Ba2+ or Fe3+ [198]. Generally,
neutralization of cationic and anionic charged within its structure [166]. spherical alginated-based microcapsules or microspheres are produced
Gelatin solidifies to form crystalline regions within the amorphous ma- by directly dripping the sodium alginate solution into CaCl2 solution
trix at room temperature [189]. Gelatin displays a thermoreversible [198]. Builders et al. [99] developed the mucinated sodium alginate
property. The chain of gelatin partially renatures, and forms interchain microcapsules for oral delivery of insulin. The efficacy of microencapsu-
and intrachain collagen-like triple-stranded helices at below 30 ºC lated insulin was examined using a diabetic rabbit model. The blood glu-
while the helices remelt and gelatin returns to its random coil configu- cose lowering effect induced by the orally administered insulin-loaded
ration at higher than 40 °C [190]. The average skin temperature ranges microparticles prepared with three parts of sodium alginate and
from 29.19 to 35.90 °C between the slightly warm or slightly cool and one part of mucin after 5 h was equal to that produced by the subcuta-
neutral conditions [191]. The crosslinked gelatin network could swell neously administered insulin solution. It was believed that mucinated
under the release solvent and allow the egress and diffusion of the sodium alginate microcapsules could be an effective alternative for
entrapped drug [192]. The polymer chain relaxation of drug loaded gel- oral delivery of insulin. Rastogi et al. [199] reported the synthesis of
atin matrices occurred and the gelatin network started to swell, causing alginate microspheres for the oral delivery of isoniazid. The isoniazid
an increase in the matrix volume and smaller pores in gelatin matrices. loaded alginate microspheres were orally administered by adult Wister
This action generated spaces between the polymer chains, resulting in rats. Gamma-scintigraphic studies were conducted to determine the
the release of the entrapped drug [192]. The free volume in a polymer location of microspheres in rat body after oral administration. The
network might be considered as a volume fraction of molecular-sized presence of microspheres could be marked in the intestinal lumen 4 h
holes which were accessible for diffusion [193]. Therefore, the gelatin- post-oral administration. Microspheres could also be detected in the in-
based system might allow the sustained release of entrapped drugs by testine after 24 h although the percent of radioactivity had significantly
attaching on the skin for topical application. The polymer chains of mi- decreased. Ishak et al. [200] investigated the performance of chitosan-
crocapsules might relax when subject to the moisture and heat generat- coated floating alginate beads for oral delivery of micronized Metroni-
ed from the skin, permitting diffusion of the entrapped drugs from the dazole (MZ) using a Helicobacter pylori (H. pylori)-induced mice
micro-pores or macromolecular network of the matrix carrier through model. The histopathological assay showed that groups receiving MZ
the stratum corneum into the epidermis. The drug might then be in the form of floating alginate beads at 10, 15 and 20 mg/kg possessed
absorbed in the capillary network before entering the peripheral circu- a better eradication of H. pylori infection in mice than that of the suspen-
lation (Fig. 7). Lam et al. [194] investigated the washing fastness of ber- sion form of free MZ. The in vivo H. pylori clearance study also revealed
berine loaded chitosan and gelatin/agar microcapsules containing that 15 mg/kg MZ loaded floating alginate beads showed 100% clear-
textile fabrics. The results revealed that gelatin/agar-based microcap- ance rate while 20 mg/kg MZ containing suspension resulted in only
sules containing the internal water phase of berberine maintained rela- 33.33%. Sandhya et al. [201] designed the glipizide mucoadhesive
tively higher amount on cotton fabrics and exhibited better antibacterial microcapsules coated with alginate and sweet potato starches
activity even after 20 washing cycles when compared with chitosan mi- which were suitable for oral drug delivery. The developed microcap-
crocapsules having the core oil phase of berberine. Therefore, gelatin- sules exhibited a slow and complete release of glipizide over a
based system would be more suitable for the delivery of topically ap- prolonged period of time and a good mucoadhesive property in the
plied medicines. in vitro wash-off test.
P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45 37

7.1.4. Cellulose-based systems amifostine. In vitro release study showed that about half of amifostine
Cellulose is a linear polysaccharide originated from natural sources was released during the first 6 h, and the drug continued to release,
including cotton, hemp, wood and other plants. It is highly crystalline with 92% of amifostine release after 12 h. The developed PLGA
due to the extensive intramolecular hydrogen bonds. β-Glucose mole- microcapsule could be a carrier for the orally active controlled-release
cules condense through β(1 → 4)-glycosidic bonds to lead to cellulose of amifostine. Tian et al. [216] prepared the PLGA microcapsules con-
[196,202]. Cellulose has been reported as a wall material of microcap- taining plasmid DNA (pDNA) against lymphocystis disease virus
sules for oral and transdermal deliveries in several studies. Prajapati (LCDV). In vivo fish model demonstrated that oral administration of
et al. [203] studied the hypoglycemic performance of alginate–sodium microcapsules containing the pDNA could induce the immune system
carboxymethyl cellulose or carbopol 934P microcapsules with gliclazide to produce the corresponding fish-anti-LCDV antibody which could be
entrapment. The in vivo diabetic albino rat assay demonstrated that the detected in the sera of Japanese flounders. The results implied that
alginate–sodium carboxymethyl cellulose microencapsulated drug had PLGA microcapsules were effective as oral carriers for the transfer of
a prolonged hypoglycemic effect (2–16 h) when compared with the pDNA. Santoyo and co-workers [217] reported the PLGA microparticles
oral administration of the free drug (0.5–10 h). Pal and Nayak [204] loaded with acyclovir against viruses of the Herpes group. The skin
reported the development of gliclazide-loaded alginate–methyl cellulose penetration study using a porcine skin model showed that there was
microcapsules with good mucoadhesion. In vivo alloxan-induced diabet- no noticeable difference between the quantity of the drug in the basal
ic albino rat model showed that significant hypoglycemic activity could epidermis with microparticles and with acyclovir suspension at 6 and
be noted in gliclazide-loaded microcapsule group after oral administra- 24 h. However, the acyclovir reservoir in the basal epidermis with
tion. Murtaza et al. [205] reported the synthesis of ethylcellulose microparticles was higher than that with the control suspension after
tabletted microparticles for oral delivery of salbutamol sulfate. The in- 88 h. This could be probably due to the controlled drug release pro-
vitro drug release test showed that an initial rapid release of salbutamol duced by the vector in the basal epidermis. The PLGA microparticles
sulfate from the tabletted microparticles was observed, followed by a could improve acyclovir topical therapy as the drug retention in
slow and prolonged drug release over 12 h. The in vivo study with the in- the basal epidermis could be increased. This group [107] further inves-
volvement of young healthy human volunteers demonstrated the bio- tigated the topical drug penetration of cidofovir loaded PLGA micropar-
availability of drug loaded microcapsules after oral administration. The ticles against herpes viruses. Although the skin penetration assay using
drug could be detected in the human plasma samples. Hu et al. [206] re- a porcine skin model revealed that the amount of drug penetration was
ported the development of clarithromycin (CLM) loaded ethyl cellulose higher with the drug solution than with microparticles after 24 h, the
microcapsules for oral drug delivery. The pharmacokinetic study showed amount of cidofovir found in the basal epidermis was much higher
that CLM dry suspension containing microcapsules did not decrease the with microparticles than with the control solution. The results sug-
bioavailability and did possess a better therapeutic performance for gested that cidofovir-loaded microparticles could increase the drug re-
prolonging the release behavior significantly in healthy volunteers. tention in the basal epidermis and reduce its penetration through the
Giandali et al. [207] showed the synthesis of vancomycin loaded treha- skin. Hrynyk et al. [108] developed the human recombinant crystalline
lose/hydroxyethylcellulose microspheres for topical drug delivery. The insulin (hRcI) loaded PLGA microcapsules for the topical wound healing
developed microspheres might keep the treated skin site dry and pro- treatment. In vitro scratch assay showed that insulin released from
vide the sustained release of vancomycin. The vancomycin loaded micro- PLGA microspheres over 23 days stimulated a rapid cell migration fol-
spheres were suggested to be useful for topical applications on the lowing an induced scratch as fresh insulin, suggesting a high level of
potential microbial contamination on skin wounds when there were ex- sustained bioactivity.
tensive injuries in skin and damages in skin barriers.
7.2.2. Poly (vinyl alcohol) (PVA)-based systems
7.2. Synthetic polymer-based systems for drug microencapsulation PVA is a water-soluble synthetic polymer with biocompatible, biode-
gradable, nontoxic and excellent mechanical properties [218,219].
7.2.1. Poly(L-lactic acid) (PLA)- and poly(lactic-coglycolic acid) (PLGA)- Crosslinked PVA is able to swell in aqueous conditions to offer a
based systems controlled delivery of drugs [220]. Kaity et al. [221] synthesized a
PLA and PLGA have been used for drug microencapsulation due to novel interpenetrating polymer network microspheres of locust bean
their biodegradable, biocompatible and non-toxic properties, and they gum (LBG) and PVA for oral controlled release of buflomedil HCl using
have been approved by the Food and Drug Administration for human glutaraldehyde as the crosslinking agent. An increase in the amount of
use [208,209]. PLA is linear aliphatic polyester synthesized from lactic PVA and glutaraldehyde decreased the wake uptake and drug release
acid monomers which are obtained by the fermentation of biomass, because of the low water uptake capacity of PVA and the increased
such as corn or wheat, or waste products, such as whey or molasses rigidity of microcapsule network by glutaraldehyde. The developed
[210]. However, the hydrophily of PLA is unsatisfactory, resulting in a microspheres could be a potential candidate for providing a controlled
slow decomposition rate [211]. PLGA is depolymerized in the presence release of drugs with short half-life and high water solubility. Jain and
of water. The hydrolysis reaction cleaves the ester bonds of polymer coworkers [222] developed the glutaraldehyde crosslinked Lepidium
chains [212]. PLGA allows the controlled drug release from its pas- sativum (Ls) and PVA interpenetrating polymer network microspheres
sive degradation in aqueous environments such as living tissues to provide a controlled drug release to improve the bioavailability
[85,212,213]. PLA and PLGA have been reported for micro-particu- and half-life of simvastatin after oral administration. In vitro release
late oral and topical drug deliveries. Ma et al. [214] developed the study showed that an increase in the amount of glutaraldehyde or
orally administered PLA microcapsules in order to protect insulin PVA could lead to a denser polymer network of microspheres which
from proteolytic enzymes in the gastrointestinal tract. The in vivo di- resulted in a slower drug release from the polymeric matrix. This
abetic rat model showed that the insulin loaded PLA microcapsules group [223] further produced the interpenetrating polymer network
were capable of lowering the blood glucose level. Sixty-eight percent of gum ghatti (Gg) and PVA microspheres using glutaraldehyde as a
of diabetic rat blood glucose level was reduced by 68.5 ± 11.7% and crosslinker for the oral controlled release of ranitidine HCl. The results
21% of diabetic rat blood glucose level was decreased by 39.7 ± 6.6%. showed that an increase in the amount of glutaraldehyde or PVA
The results suggested that PLA microcapsule could be used as a car- resulted in a higher mucoadhesivity and a slower drug release.
rier to protect insulin from proteolytic degradation in the gastroin- Microspheres with a higher proportion of Gg:PVA (1:4) and a higher
testinal tract and ensure the bioactivity of insulin after the passage amount of glutaraldehyde (3.5 mL) had the highest mucoadhesivity
through the intestinal tract. Mandal et al. [215] designed the PLGA 82.33 ± 0.57% in the in vitro wash off test and the slower drug release
biodegradable microcapsules for the potential oral delivery of in the release study, when compared with microspheres prepared
38 P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45

with Gg:PVA (1:2) and 1.5 mL glutaraldehyde, with the lowest 7.3. Multiple microemulsion systems for drug microencapsulation
mucoadhesivity 62.66 ± 0.57% and the faster drug release.
A water-in-oil-in-water (w1/o/w2) multiple emulsion system con-
7.2.3. Poly(ɛ-caprolactone) (PCL)-based systems sists of the internal (w1) and external (w2) phases, which are chemically
PCL is considered as a promising candidate for controlled drug alike, as well as an intermediate immiscible (o) phase which physically
delivery due to its biodegradable, biocompatible, non-toxic characteris- separates the two alike phases [235]. The formation of a stable multiple
tics [76,224,225]. The high crystallinity and hydrophobicity of PCL emulsion involves a two-step emulsification process, where in the first
result in a relatively slow biodegradation rate and an unsuitable micro- step a water-in-oil emulsion (w1/o) is initially formed, which is
environment for tissue growth [226]. Benoit et al. [227] reported the then re-emulsified in an aqueous phase (w2) with a combination of
development of bovine serum albumin loaded PCL microparticles emulsifiers and/or stabilizing agents [236]. Therapeutic drugs could be
which were suitable for oral vaccine delivery. The study confirmed encapsulated in the internal (w1) phase and the intermediate immisci-
that the protein could maintain unchanged without the presence of ag- ble (o) phase of the multiple emulsion using microencapsulation
gregates during the encapsulation process. Barboza et al. [228] devel- technology for both oral and topical drug deliveries. The w/o/w
oped the PCL and poly(3-hydroxybutyrate-co-3-22 hydroxyvalerate) micro-particulate system could be developed by a micro-emulsion. A
(PHBV) microparticles for the oral controlled release of manidipine microemulsion is defined as a single optically isotropic structured solu-
dihydrochloride (MAN). The in vivo rat model revealed that MAN load- tion containing water, oil and surfactant [237]. By applying the
ed PCL microparticles could reduce the mean arterial pressure variation microemulsion vehicles, both hydrophilic and oil-soluble agents could
up to 24 h after phenylephrine injections when compared with the pure be solubilized so as to achieve a cooperative function for a specific ther-
drug and MAN loaded PHBV microparticles. The orally administered apeutic use [41]. Previous studies reported the synthesis of w/o/w
PCL microparticles containing MAN were suggested to promote a microemulsion systems. Kawakatsu et al. [238] reported the develop-
long-lasting antihypertensive effect. ment of w/o/w emulsion for producing microcapsules. In the w/o sys-
tem, pectin, as a polysaccharide, was used as an internal water phase
7.2.4. Poly(methyl methacrylate) (PMAA)-based systems while oleic acid or triolein containing Span 20, Span 80 or TGPR were
PMAA is a biocompatible ionizable hydrophilic polymer [229–231]. used as an oil phase. The w/o/w emulsion was then produced by feeding
Crosslinked PMAA network is capable of swelling in water and its swell- w/o emulsion through the micro-channels into an external water phase
ing behavior highly depends on the pH value because of the ionization/ containing 3 wt.% of Tween 20. Foster et al. [239] designed the w/o/w
deionization of the carboxylic acid groups. The \COOH groups are not emulsion for microcapsule formation which was suggested to be
ionized and the PMAA network remains at its collapsed state at low applied to oral drug delivery, cell encapsulation, protein evolution
pH of less than 5.5, whereas the \COOH groups are ionized and the and gene therapy. In this system, a biocompatible and biodegradable
charged COO− groups repulse each other at high pH of 5.5–7.4, resulting amphiphilic diblock copolymer poly(ethylene glycol)-b-poly(c-methyl
in the swelling of PMAA [231,232]. Eudragit® L100-55 is an anionic e-caprolactone) was used as the internal water phase; volatile oil was
methacrylic acid–ethyl acrylate copolymer which provides a coating of applied as the oil phase; and PVA was employed as the outer water
microparticles with gastro-resistant property. Kovacs-Nolan et al. phase. Cai et al. [240] developed the polystyrene hollow spheres using
[233] reported the development of anti-VP8 IgY loaded Eudragit® the w/o/w multiple emulsions. Sodium lauryl sulfate, as an internal
L100-55 microcapsules to increase a protective efficacy under gastroin- water phase, was suspended in polystyrene dissolved benzene and
testinal conditions after oral administration. The in vitro stability study 1,2-dichloroethane. The w/o emulsion was then poured to the PVA
demonstrated that a significantly more activity was preserved by micro- which was used as the outer water phase. Liu et al. [241] reported the
encapsulated IgY (a 35%w/w enteric polymer coating resulted in the re- synthesis of PLA/PLGA microcapsules loaded with recombinant human
tention of greater than 95% of IgY activity after 6 h of incubation in SGF) insulin using a w1/o/w2 multiple emulsion. The inner water phase
than non-encapsulated IgY, with very little retention of IgY activity (w1) consisted of recombinant human insulin while the oil phase (o) in-
(a significant decrease (86%) in IgY activity after 30 min of SGF expo- volved a mixture solvent of dichloromethane (DCM) and toluene con-
sure). The in vivo stability test of the IgY using a weaned pig model taining PLA/PLGA and Arlacel 83. The outer water phase (w2) was
also revealed that there was a positive correlation (r = 0.996) between composed of PVA and sodium chloride which were dissolved in water.
the IgY activity and the amount of IgY in different regions of the gastro- The multiple microemulsion systems possess the advantages for both
intestinal tract for the microencapsulated IgY. However, the amount of oral and topical drug deliveries. Oral drug delivery using the multiple
non-encapsulated IgY in the gastrointestinal tract did not correlate as microemulsion reduces the transient drug overload, resulting in a de-
well with IgY activity (r = 0.615) and the result showed high quantities crease in local adverse side effects [242]. Microemulsion declines the
of IgY yet low IgY activity, suggesting that the antibodies were present, first-pass metabolism and remains the plasma drug level for a
but they lost their activity. The results demonstrated that the prolonged period of time [243]. Microemulsion also possesses some
microencapsulated IgY with a pH-sensitive methacrylic acid copolymer benefits for transdermal drug delivery. An increase in drug concentra-
(Eudragit® L100-55) could protect the antibodies from gastric tion in the skin is achieved due to a large amount of drug incorporated
inactivationto promote an effective control of HRV infection. in the formulation. An increased thermodynamic stability of the drug
Eudragit® L30 D-55 is an aqueous dispersion of an anionic may enhance its partitioning into the skin. The components of
polymethacrylate and it is able to provide the microcapsules with microemulsion may act as permeation enhancers to decrease the diffu-
gastro-resistant effects. G. Año et al. [234] synthesized the vibrio sional barrier of the stratum corneum, leading to an increase in the per-
cholerae C7258 inactivated strain (VC) embedded Eudragit® L30 meation rate of drug through the skin [244,245]. Cournarie et al. [246]
D-55 microcapsules. The results obtained from the immunogenicity compared the insulin loaded w/o/w multiple microemulsion prepared
assay of microparticles using an oral inoculation adult rat model with either Miglyol® 810N or fish oil using a two-step process. The mul-
showed that the oral administration of 3.5 mg of microencapsulated tiple microemulsion consisted of an inner aqueous phase of 71% of an
VC achieved an appropriate protection of the bacteria which induced aqueous solution of insulin (2.8 mg/mL) containing 0.18% of NaCl; an
the production of high levels of vibriocidal titers. This suggested the oil phase of 25% of oil (either Miglyol® 810N or fish oil), 4% of Abil®
possibility of using lower doses of microencapsulated VC to achieve a EM-90; and an external aqueous phase of Arlatone® F127G (1%). This
suitable immune response. However, VC alone at 3.5 mg did not in- study suggested that the combination of Miglyol® 810N and fish oil
duce an immune response. The developed microcapsules could was proposed to prepare the multiple microemulsion in order to obtain
offer a low cost and easily produced oral vaccine, which would be the benefits of Miglyol® 810N (small globules formation and higher sta-
helpful to be administered to young children. bility) and fish oil (beneficial therapeutic effects) for oral administration.
P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45 39

Shahiwala et al. [247] developed the squalane oil-containing w/o/w mul- DNAzymes. This study concluded that the w/o/w multiple emulsion
tiple emulsion for the oral immunization of ovalbumin (OVA). Oral im- was helpful to transport the DNAzyme across the skin and into epider-
munizations of squalane oil-containing w/o/w multiple emulsion mal cells of the atopic dermatitis affected skin. An interesting w1/o/w2
resulted in a much higher systemic IgG and mucosal IgA responses multiple microemulsion system has been developed for the potential
when compared with the nasally treated groups. The results revealed applications of oral and transdermal drug deliveries (Lam's unpublished
that squalane oil-containing w/o/w multiple emulsion formulations data). Gelatin was used as an internal water phase (w1) containing a bio-
could significantly improve the local and systemic immune re- medical agent. The oil phase (o) was a vegetable oil while the outer
sponses after oral administration. The developed system could be a phase (w2) was chitosan. Fig. 8 shows this w1/o/w2 microemulsion sys-
better alternative to the mucosal delivery of prophylactic and therapeu- tem. The developed emulsion system is speculated to be a drug carrier
tic vaccines. Paul et al. [248] developed the w/o/w multiple emulsions for both oral and topical applications for specific disease treatments.
for acyclovir entrapment to enhance its oral bioavailability. The in vivo
rat model showed that an extended drug release was obtained from 8. Challenges of drug microencapsulation in real life
w/o/w multiple emulsions, resulting in a better oral bioavailability
when compared with the drug solution. Tirnaksiz and Kalsin [249] de- Drug microencapsulation possesses a significant potential for phar-
veloped the formulation of topical w/o/w multiple microemulsions maceutical and therapeutic fields in future as it provides the sustained
with the globule size ranging from 20 to 37 μm. The inner water phase and controlled release of pharmaceutical agents for various medical
included 0.03% sodium chloride and 1.5% caffeine in distilled water, applications. However, microencapsulation technology still faces the
the oil phase involved 30% liquid paraffin and 2 or 4% lipophilic surfac- existing obstacles and limitations in drug deliveries. This results in a dis-
tant (Abil®EM90), and the outer water phase contained 1, 2 or 4% tance between drug microencapsulation and clinical practice in real life.
Tetronic®908 in distilled water. An initial rapid release of caffeine The challenge involves the scale-up of drug microencapsulation process
followed by a much slower rate of drug release could be shown. The op- to achieve the high reproducibility of the microencapsulated drug
timal multiple emulsion involving Tetronic®908 (1%) in the outer aque- formulations. Technical, quality, ethical and economic issues should be
ous phase and Abil®EM90 (2%) in the oil phase was considered as a considered when the mass production of pharmaceuticals using micro-
potential dosage form of prolonged drug release. Schmidts et al. [250] encapsulation technology is implemented. Technical considerations
reported the development of a topically used w/o emulsion, w/o/w may include the features and requirements of a microencapsulated
emulsion, multiple emulsion and submicron emulsion for the pharmaceutical product that should be specified and identified before
DNAzymes delivery to treat inflammatory diseases such as atopic der- the design of a drug formulation. For examples, the microencapsulated
matitis. The w/o emulsion involved the aqueous phase (5 wt.% glycerol, protein should be planned to prolong the drug bioactivity after oral ad-
0.3 wt.% MgSO4, 0.14 wt.% potassium sorbate, 0.07 wt.% citric acid and ministration using a low permeable and biocompatible polymeric ma-
63.9 wt.% distilled water) and the oil phase (3.0 wt.% triglycerol trix for drug entrapment. Precise characterization of drug loaded
diisostearate, 1.2 wt.% isopropyl palmitate, 1.2 wt.% Cetostearyl microcapsules is fundamental to the development of a pharmaceutical
Isononanoate, 0.2 wt.% soy lecithin and 24.6 wt.% hydrophobic basis formulation for the disease treatment. For the therapeutic applications,
gel). For the w/o/w emulsion, the inner aqueous phase consisted of the physical, chemical and therapeutic properties of microcapsules
0.065 M MgSO4 solution containing the DNAzyme, the oil phase should be carefully examined and identified. Drug entrapment efficien-
contained 15.8 wt.% heavy paraffin oil, 4.0 wt.% sorbitan oleate and cy of microcapsules is closely related to the drug doses and therefore it
0.2 wt.% soy lecithin, and the outer aqueous phase was composed of should be cautiously controlled to avoid an overdose administration of
1.0 wt.% Steareth-20 and 58.7 wt.% preserved water. The multiple emul- the drug. Particle size of microcapsules could alter the total surface
sion was produced by mixing the appropriate quantities of the compo- area for the drug release. The stability of microcapsules should be ma-
nents and the DNAzyme at room temperature. The oil phase contained nipulated in order to produce the microencapsulated pharmaceuticals
7.20 wt.% Labrasol®, 4.80 wt.% Plurol Oleique®, 2.50 wt.% cetostearyl with a clinically acceptable shelf life. The materials involved in the mi-
isononanoate and 2.50 wt.% isopropyl palmitate, and the water phase crocapsule formation should be standardized in terms of chemical com-
was composed of 2.49 wt.% glycerol, 16.60 wt.% propylene glycol, positions, purification and reaction situations so as to achieve the
0.30 wt.% MgSO4 and 63.21 wt.% distilled water. For the submicron biosafety and efficiency requirements consistently and minimize the
emulsion, the water phase (a blend of Oleth-3 and Oleth-10 in total product variability in different laboratories [251,252]. Additionally, the
6.0 wt.%, 3.0 wt.% glycerol, 0.3 wt.% MgSO4 and 75.3 wt.% preserved
water) and oil phase (5.0 wt.% Coco-Caprylate/Caprate, 5.0 wt.%
cetearyl isononanoate, 5.0 wt.% Ethyl oleate) were heated separately
to 70 °C and then mixed together. Oleth-3 and Oleth-10 were used to
adjust the HLB values of 10, 11 and 12 for three different submicron
emulsions. The stability test showed that w/o/w emulsion and w/o
emulsion had the higher DNAzyme stability over 3 months as compared
with submicron emulsion and multiple emulsion. The skin penetration
studies using the porcine skin model showed that the submicron emul-
sion (HLB 12) exhibited the highest DNAzyme penetration for both in-
tact (untreated) and impaired (with a simulated impaired barrier of
atopic dermatitis skin) skin after 24 h, followed by the multiple emul-
sion with the second highest DNAzyme uptake. DNAzyme uptake from
submicron emulsion to the skin revealed that components such as
ethyl oleate, interacted with the stratum corneum, resulting in an in-
crease in DNAzyme penetration as compared to skin with impaired bar-
rier (a reduced stratum corneum). The low DNAzyme uptake by w/o/w
emulsion and w/o emulsion might be associated with the higher stabil-
ity which delayed the release of the entrapped DNAzyme. However, the
oily membrane of w/o/w multiple emulsion broke down under shear
during topical application, resulting in the successful release and skin
penetration of DNAzyme, as well as the higher cellular uptake of Fig. 8. A w1/o/w2 multiple microemulsion system.
40 P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45

quality control should be necessary to produce the microencapsulated 10. Conclusion


medicine with an acceptable quality. Ethical approval of a pharmaceuti-
cal product should be obtained to demonstrate the safe use of drug load- Recent advances in microencapsulation systems for the applications
ed microcapsules as being supported by the results of animal of oral and transdermal drug deliveries are summarized in this review.
experiments and clinical trials. The operational procedures of drug mi- The use of micro-particulate systems for producing biopharmaceuticals
croencapsulation should be simplified to increase the efficiency of pro- and micro-medicines has shown to have a great potential in pharma-
duction, and thus reducing the production time and cost. All of these ceutical and medical industries. The desired formulations should protect
steps are fundamental to the application of drug microencapsulation the drugs under unstable biological environments, such as drug degra-
technology for clinical therapies in the real life. dation induced by the gastrointestinal tract and first-pass liver effects
after oral administration, overcome the low skin permeability when
9. Commercially available microencapsulated pharmaceuticals for the drugs are administered through transdermal route, and promote a
oral and transdermal drug deliveries prolonged drug delivery to the targeted body sites. Micro-particulate
systems for oral and transdermal drug deliveries are believed to over-
Although the challenges and limitations of drug microencapsulation come the flaws in the conventional drug dosage forms They also leave
exist, there are some successful microencapsulation products which an alternative administration route to improve the drug release profile,
have been commercially available in the market. For the microencapsu- and enhance the bioavailability and stability of the drug dosages. In fact,
lated oral drug delivery systems, Bayer Inc. has developed the CIPRO® the synthesis and development of microencapsulation systems for drug
Oral Suspension which consists of ciprofloxacin microcapsules and delivery are mature. However, the actual applications and usages of
strawberry flavored diluent medium for oral administration to treat these microencapsulation systems remain to be limited. Although
the microbial infections [253]. Micropharma Ltd. has launched the some progress has been made in recent studies, few previous studies in-
Cardioviva™ that contains the alginate-poly-L-lysine-alginate microen- vestigated the possible biological effects and therapeutic activities from
capsulated probiotic Lactobacillus reuteri NCIMB 30242 for oral adminis- in vitro and in vivo models. The next challenge will be the bridging of the
tration. The clinical study reported that the Cardioviva™ could synthesis and potential applications of medicated microencapsulation
effectively lower the cholesterol level after a 6 week treatment period systems.
[254]. Ther-Rx Corp., a branded-drug subsidiary from KV Pharmaceuti-
cal Co., has marketed the Micro-K® Extencaps® which is an oral formu-
Acknowledgment
lation of microencapsulated potassium chloride for the treatment of
hypokalemia [255]. Mayne Pharma International has developed the
Parts of this work were extracted from P.L. Lam's thesis. The authors
taste masked Paracetamol (Nopap™) powder and tablets which can
would like to acknowledge the support of the Innovation Technology
mark the unpleasant taste of the drug, enhance the drug stability and
Commission and Research Grants Council, The Hong Kong Polytechnic
provide a sustained drug release after oral administration [256].
University. Prof. R. Gambari is supported by AIRC.
Novartis Int. AG has invented the Sandimmun Neoral®, an oral formula-
tion of Cyclosporin A, using the microemulsion technology for the treat-
ment of rheumatoid arthritis. The clinical study reported that the References
microemulsion formulation improved the drug bioavailability, resulting
in a reduced requirement for increased doses over time [257]. Roche has [1] T.A. Al-Hilal, F. Alam, Y. Byun, Oral drug delivery systems using chemical
conjugates or physical complexes, Adv. Drug Deliv. Rev. 65 (2013) 845–864.
marketed the Fortovase® microemulsion formulation for the oral deliv- [2] M. Foldvari, Non-invasive administration of drugs through the skin: challenges in
ery of Saquinavir and Abbott Laboratories has also developed the delivery system design, Pharm. Sci. Technol. Today 3 (2000) 417–425.
Norvir®, an oral microemulsion dosage of ritonavir for the treatment [3] K. Thanki, R.P. Gangwal, A.T. Sangamwar, S. Jain, Oral delivery of anticancer drugs:
challenges and opportunities, J. Control. Release 170 (2013) 15–40.
of Human Immunodeficiency Virus (HIV) [258]. For the microencapsu- [4] J. Renukuntla, A.D. Vadlapudi, A. Patel, S.H.S. Boddu, A.K. Mitra, Approaches for
lated topical drug delivery systems, Tagra biotechnologies Ltd. has syn- enhancing oral bioavailability of peptides and proteins, Int. J. Pharm. 447 (2013)
thesized the Tagravit™ microcapsules for topical delivery of vitamins 75–93.
[5] G. Eichenbaum, J. Zhou, A. De Smedt, S. De Jonghe, A. Looszova, T. Arien, F. Van
(A, E and F) onto the skin. Tagravit™ microcapsules significantly im- Goethem, I. Vervoort, U. Shukla, L. Lammens, Methods to evaluate and improve
prove the stability of vitamins by protecting the vitamins from the ex- the injection site tolerability of intravenous formulations prior to first-in-human
ternal environments [259]. DS Laboratories, Inc. has invented the testing, J. Pharmacol. Toxicol. Methods 68 (2013) 394–406.
[6] G.Z. Martins, C.R.F. Souza, T.J. Shankar, W.P. Oliveira, Effect of process variables on
Trioxil® (bisazulene gel) in a carrier agent of natural encapsulated gin-
fluiddynamics and adhesion efficiency during spouted bed coating of hard gelatine
seng and hamamelis extracts as well as arnica for topical delivery. capsules, Chem. Eng. Process. Process Intensif. 47 (2008) 2238–2246.
Trioxil® acts for 12 h to inhibit the Propionibacterium acnes bacteria [7] W.J. Elliott, Optimizing medication adherence in older persons with hypertension,
and fungi after applying onto the skin [260]. Table 3 shows the commer- Int. Urol. Nephrol. 35 (2003) 557–562.
[8] M.A. Krousel-Wood, A. Hyre, P. Muntner, D. Morisky, Methods to improve medica-
cially available microencapsulation products for oral and transdermal tion adherence in hypertensive patients: current status and future directions, Curr.
drug deliveries. Opin. Cardiol. 20 (2005) 296–300.

Table 3
Commercially available microencapsulation products for oral and transdermal drug deliveries.

Product name Core active ingredient Dosage route Developer/marketer Indication References

CIPRO® Ciprofloxacin Oral Bayer Inc. Microbial infection [253]


Cardioviva™ Lactobacillus reuteri NCIMB 30242 Oral Micropharma Ltd. Hypercholesterolemia [254]
Micro-K® Extencaps® Potassium chloride Oral KV Pharmaceutical Co. Hypokalemia [255]
Nopap™ Paracetamol Oral Mayne Pharma International Pain and fever [256]
Sandimmun Neoral® Cyclosporin A Oral Novartis Int. AG Rheumatoid arthritis [257]
Fortovase® Saquinavir Oral Roche HIV [258]
Norvir® Ritonavir Oral Abbott Laboratories HIV [258]
Tagravit™ Vitamin A, E and F Transdermal Tagra Biotechnologies Ltd. Aging [259]
Trioxil® ginseng and hamamelis extracts, arnica Transdermal DS Laboratories Propionibacterium acnes bacteria and fungi [260]
P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45 41

[9] Z.H. Ma, D.G. Yu, C.J. Branford-White, H.L. Nie, Z.X. Fan, L.M. Zhu, Microencapsula- [41] M.R. Prausnitz, Overcoming skin's barrier: the search for effective and user-friendly
tion of tamoxifen: application to cotton fabric, Colloids Surf. B: Biointerfaces 69 drug delivery, Diabetes Technol. Ther. 3 (2001) 233–236.
(2009) 85–90. [42] A. Kogan, N. Garti, Microemulsions as transdermal drug delivery vehicles, Adv.
[10] P.L. Lam, R.S.M. Wong, M.C.W. Yuen, K.H. Lam, R. Gambari, C.H. Chui, Biomedical Colloid Interface Sci. 123–126 (2006) 369–385.
textiles with therapeutic effects: development of berberine containing chitosan [43] M.A. Bayomi, Aqueous preparation and evaluation of albumin–chitosan micro-
microcapsules, Minerva Biotecnol. 24 (2012) 62–69. spheres containing indomethacin, Drug Dev. Ind. Pharm. 30 (2004) 329–339.
[11] P.L. Lam, K.K.H. Lee, S.H.L. Kok, G.Y.M. Cheng, X.M. Tao, D.K.P. Hau, M.C.W. Yuen, [44] M. Saravenan, K. Bhaskar, G.S. Rao, M.D. Dhanaraju, Ibuprofen-loaded
K.H. Lam, R. Gambari, C.H. Chui, R.S.M. Wong, Development of formaldehyde-free ethylcellulose/polystyrene microspheres: an approach to get prolonged drug
agar/gelatin microcapsules containing berberine HCl and gallic acid and their top- release with reduced burst effect and low ethylcellulose content, J. Microencapsul.
ical and oral applications, Soft Matter 8 (2012) 5027–5037. 20 (2003) 289–302.
[12] P.L. Lam, K.K.H. Lee, R.S.M. Wong, G.Y.M. Cheng, S.Y. Cheng, M.C.W. Yuen, K.H. Lam, [45] P. Valot, M. Baba, J.M. Nedelec, N. Sintes-Zydowicz, Effects of process parameters
R. Gambari, S.H.L. Kok, C.H. Chui, Development of hydrocortisone succinic acid/and on the properties of biocompatible Ibuprofen-loaded microcapsules, Int. J. Pharm.
5-fluorouracil/chitosan microcapsules for oral and topical drug deliveries, Bioorg. 369 (2009) 53–63.
Med. Chem. Lett. 22 (2012) 3213–3218. [46] R. Pandey, A. Zahoor, S. Sharma, G.K. Khuller, Nanoparticle encapsulated antituber-
[13] P.L. Lam, M.C.W. Yuen, C.W. Kan, R.S.M. Wong, G.Y.M. Cheng, K.H. Lam, R. Gambari, cular drugs as a potential oral drug delivery system against murine tuberculosis,
S.H.L. Kok, C.H. Chui, Development of calendula oil/chitosan microcapsules and Tuberculosis 83 (2003) 373–378.
their biological safety evaluation, Aust. J. Chem. 65 (2012) 72–80. [47] M. Petitti, A.A. Barresi, M. Vanni, Controlled release of vancomycin from PCL micro-
[14] P.L. Lam, R. Gambari, J. Yip, M.C.W. Yuen, K.H. Lam, R.S.M. Wong, X.W. Wang, J.C.O. capsules for an ophthalmic application, Chem. Eng. Res. Des. 87 (2009) 859–866.
Tang, S.H.L. Kok, C.H. Chui, Development of phyllanthin containing microcapsules [48] Z. Mao, L. Ma, C. GaoT, J. Shen, Preformed microcapsules for loading and sustained
and their improved biological activity towards skin cells and staphylococcus release of ciprofloxacin hydrochloride, J. Control. Release 104 (2005) 193–202.
aureus, Bioorg. Med. Chem. Lett. 22 (2012) 468–471. [49] M. Song, N. Li, S. Sun, L.R. Tiedt, W. Liebenberg, M.M. deVilliers, Effect of viscosity
[15] K.H. Lam, S.Y. Cheng, P.L. Lam, C.W.M. Yuen, S.M.R. Wong, F.Y. Lau, B.S.P. Lai, R. and concentration of wall former, emulsifier and pore-Inducer on the properties
Gambari, C.H. Chui, Microencapsulation: past, present and future, Minerva of amoxicillin microcapsules prepared by emulsion solvent evaporation, II,
Biotecnol. 22 (2010) 23–28. Farmaco 60 (2005) 261–267.
[16] S.S. Bansode, S.K. Banarjee, D.D. Gaikwad, S.L. Jadhav, R.M. Thorat, Microencapsulation: [50] Z. Zhang, Y. Zhu, X. Yang, C. Li, Preparation of azithromycin microcapsules by a
a review, Int. J. Pharm. Sci. Rev. Res. 1 (2010) 38–43. layer-by-layer self-assembly approach and release behaviors of azithromycin,
[17] A.K. Aggarwal, A. Dayal, N. Kumar, Microencapsulation process and applications in Colloids Surf. A Physicochem. Eng. Asp. 362 (2010) 135–139.
textile processing, Colourage 45 (1998) 15–24. [51] D. Sharma, T.P. Chelvi, J. Kaur, K. Chakravorty, T.K. De, A. Maitra, R. Ralhan, Novel
[18] In: T. Tadros (Ed.), Encyclopedia of Colloid and Interface Science, Springer-Verlag Taxol formulation: polyvinylpyrrolidone nanoparticle-encapsulated Taxol for
Berlin Heidelberg, UK, 2013. drug delivery in cancer therapy, Oncol. Res. 8 (1996) 281–286.
[19] H.J. Huang, X.D. Chen, W.K. Yuan, Microencapsulation based on emulsification for [52] B. Nuijen, M. Bouma, J.H. Schellens, J.H. Beijnen, Progress in development of
producing pharmaceutical products: a literature review, Dev. Chem. Eng. Miner. alternative pharmaceutical formulation of taxanes, Invest. New Drugs 19 (2001)
Process. 14 (2006) 515–544. 143–153.
[20] A. Lamprecht, R. Bodmeier, Microencapsulation, Ullmann's Encyclopedia of [53] Q. Zhao, S. Zhang, W. Tong, C. Gao, J. Shen, Polyelectrolyte microcapsules templated
Industrial Chemistry, Wiley-VCH, Weinheim, 2010. on poly(styrene sulfonate)-doped CaCO3 particles for loading and sustained
[21] P.B. Deasy, Microencapsulation and Related Drug Processes, Marcel Dekker, New release of daunorubicin and doxorubicin, Eur. Polym. J. 42 (2006) 3341–3351.
York, 1984. [54] L. Huang, W. Sui, Y. Wang, Q. Jiao, Preparation of chitosan/chondroitin sulfate
[22] F. Nazzaro, P. Orlando, F. Fratianni, R. Coppola, Microencapsulation in food science complex microcapsules and application in controlled release of 5-fluorouracil,
and biotechnology, Curr. Opin. Biotechnol. 2 (2012) 182–186. Carbohydr. Polym. 80 (2010) 168–173.
[23] M.N. Singh, K.S.Y. Hemant, M. Ram, H.G. Shivakumar, Microencapsulation: a prom- [55] X. Liu, C. Gao, J. Shen, H. Möhwald, Multilayer microcapsules as anti-cancer drug
ising technique for controlled drug delivery, Eur. J. Pharm. Sci. 5 (2010) 65–77. delivery vehicle: deposition, sustained release, and in vitro bioactivity, Macromol.
[24] J.F. Pinto, Site-specific drug delivery systems within the gastro-intestinal tract: Biosci. 5 (2005) 1209–1219.
from the mouth to the colon, Int. J. Pharm. 395 (2010) 44–52. [56] A.M. Rokstad, B.I. Gustafsson, T. Espevik, I. Bakke, R. Pfragner, B. Svejda, I.M. Modlin,
[25] J. Wojtacki, G. Rolka-Stempniewicz, M. Grzegorczyk, Breast cancer patients prefer- M. Kidd, Microencapsulation of small intestinal neuroendocrine neoplasm cells for
ences for oral versus intravenous second-line anticancer therapy, Eur. J. Cancer tumor model studies, Cancer Sci. 103 (2012) 1230–1237.
Suppl. 4 (2006) 159–160. [57] G.W. Vandenberg, C. Drolet, S.L. Scott, J. De la Noűe, Factors affecting protein
[26] M.M. Borner, P. Schoffski, R. de Wit, F. Caponigro, G. Comella, A. Sulkes, G. Greim, release from alginate–chitosan coacervate microcapsules during production and
G.J. Peters, K. van der Born, J. Wanders, R.F. de Boer, C. Martin, P. Fumoleau, Patient gastric/intestinal simulation, J. Control. Release 77 (2001) 297–307.
preference and pharmacokinetics of oral modulated UFT versus intravenous fluoro- [58] C.H. Zheng, J.Q. Gao, Y.P. Zhang, W.Q. Liang, A protein delivery system:
uracil and leucovoin: a randomized crossover trial in advanced colorectal cancer, biodegradable alginate–chitosan–poly(lactic-co-glycolic acid) (PLGA) composite
Eur. J. Cancer 39 (2002) 349–358. microspheres, Biochem. Biophys. Res. Commun. 323 (2004) 1321–1327.
[27] L.C. DeVane, Principles of pharmacokinetics and pharmacodynamics, in: A.F. [59] J.P. McQuilling, J. Arenas-Herrera, C. Childers, R.A. Pareta, O. Khanna, B. Jiang, E.M.
Schatzberg, C.B. Nemeroff (Eds.), The American Psychiatric Publishing Textbook Brey, A.C. Farney, E.C. Opara, New alginate microcapsule system for angiogenic
of Psychopharmacology, Third ed., American Psychiatric Pub, Washington, DC, protein delivery and immunoisolation of islets for transplantation in the rat
2004, pp. 181–200. omentum pouch, Transplant. Proc. 43 (2011) 3262–3264.
[28] J. Renukuntlaa, A.D. Vadlapudi, A. Patel, S.H.S. Boddu, A.K. Mitrab, Approaches for en- [60] H. Hamishehkar, J. Emami, A.R. Najafabadi, K. Gilani, M. Minaiyan, H. Mahdavi, A.
hancing oral bioavailability of peptides and proteins, Int. J. Pharm. 447 (2013) 75–93. Nokhodchi, Effect of carrier morphology and surface characteristics on the
[29] M. Davenport, Alternative routes of drug administration — advantages and disad- development of respirable PLGA microcapsules for sustained-release pulmonary
vantages (Subject Review), Pediatrics 101 (1998) 730–731. delivery of insulin, Int. J. Pharm. 389 (2010) 74–85.
[30] T.B. Patel, Recent trends in novel drug delivery system, Latest Rev. 5 (2007) 1–10. [61] M.L. De Temmerman, J. Rejman, R.E. Vandenbroucke, S. De Koker, C. Libert, J.
[31] J.A. Fix, Oral controlled release technology for peptides: status and future pros- Grooten, J. Demeester, B. Gander, S.C. De Smedt, Polyelectrolyte LbL microcapsules
pects, Pharm. Res. 13 (1996) 1760–1764. versus PLGA microparticles for immunization with a protein antigen, J. Control.
[32] V.H.L. Lee, D.K. Satish, M.G. George, R. Werner, Oral route of protein and peptide Release 158 (2012) 233–239.
drug delivery, in: Vincent H.L. Lee (Ed.), Protein and Peptide Drug Delivery: Oral [62] X.Y. Shi, T.W. Tan, Preparation of chitosan/ethylcellulose complex microcapsule and
Approaches, Marcel Dekker, New York, NY, 1991, pp. 691–738. its application in controlled release of vitamin D2, Biomaterials 23 (2002) 4469–4473.
[33] K. Golla, C. Bhaskar, F. Ahmed, A.K. Kondapi, A target-specific oral formulation of [63] S.Y. Cheng, M.C.W. Yuen, C.W. Kan, K.K.L. Cheuk, C.H. Chui, K.H. Lam, Cosmetic tex-
doxorubicin–protein nanoparticles: efficacy and safety in hepatocellular cancer, tiles with biological benefits: gelatin microcapsules containing Vitamin C, Int. J.
J. Cancer 4 (2013) 644–652. Mol. Med. 24 (2009) 411–419.
[34] I. Kuppens, T.M. Bosch, M.J. van Maanen, H. Rosing, A. Fitzpatrick, J.H. Beijnen, [64] Y.L. Xie, H.M. Zhou, X.H. Liang, B.S. He, X.X. Han, Study on the morphology, particle
J.H.M. Schellens, Oral bioavailability of docetaxel in combination with OC144-093 size and thermal properties of vitamin A microencapsulated by starch
(ONT-093), Cancer Chemother. Pharmacol. 55 (2005) 72–78. octenylsucciniate, Agric. Sci. China 9 (2010) 1058–1064.
[35] S. Peltier, J.M. Oger, F. Lagarce, W. Couet, J.P. Benoît, Enhanced oral paclitaxel bio- [65] Y. Wang, Z. Lu, H. Wu, F. Lv, Study on the antibiotic activity of microcapsule
availability after administration of paclitaxel-loaded lipid nanocapsules, Pharm. curcumin against foodborne pathogens, Int. J. Food Microbiol. 136 (2009) 71–74.
Res. 23 (2006) 1243–1250. [66] M.S. Uddin, M.N.A. Hawlader, H.J. Zhu, Microencapsulation of ascorbic acid: effect of
[36] S.C. Shin, J.S. Choi, X. Li, Enhanced bioavailability of tamoxifen after oral administration process variables on product characteristics, J. Microencapsul. 18 (2001) 199–209.
of tamoxifen with quercetin in rats, Int. J. Pharm. 313 (2006) 144–149. [67] R.K. Verma, S. Garg, Current status of drug delivery technologies and future
[37] E.I.-S. Khafagy, M. Morishita, Y. Onuki, K. Takayama, Current challenges in directions, Pharm. Technol. On-Line 25 (2001) 1–14.
non-invasive insulin delivery systems: a comparative review, Adv. Drug Deliv. [68] X. Hu, Y. Li, E. Zhang, X. Wang, M. Xing, Q. Wang, J. Lei, H. Huang, Preparation and
Rev. 59 (2007) 1521–1546. evaluation of orally disintegrating tablets containing taste-masked microcapsules
[38] M.R. Prausnitz, S. Mitragotri, R. Langer, Current status and future potential of of berberine hydrochloride, AAPS PharmSciTech 14 (2013) 29–37.
transdermal drug delivery, Nat. Rev. Drug Discov. 3 (2004) 115–124. [69] J. Zheng, X. Yue, Z. Dai, Y. Wang, S. Liu, X. Yan, Novel iron–polysaccharide
[39] B.J. Thomas, B.C. Finnin, The transdermal revolution, Drug Discov. Today 9 (2004) multilayered microcapsules for controlled insulin release, Acta Biomater. 5
697–703. (2009) 1499–1507.
[40] T.W. Prow, J.E. Grice, L.L. Lin, R. Faye, M. Butler, W. Becker, E.M.T. Wurm, C. Yoong, [70] C. Wang, W. Ye, Y. Zheng, X. Liu, Z. Tong, Fabrication of drug-loaded biodegradable
T.A. Robertson, H.P. Soyer, M.S. Roberts, Nanoparticles and microparticles for skin microcapsules for controlled release by combination of solvent evaporation and
drug delivery, Adv. Drug Deliv. Rev. 63 (2011) 470–491. layer-by-layer self-assembly, Int. J. Pharm. 338 (2007) 165–173.
42 P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45

[71] G. Orive, M. De Castro, S. Ponce, R.M. Hernandez, A.R. Gascon, M. Bosch, J. Alberch, [100] J. Zhang, Q. Tang, X. Xu, N. Li, Development and evaluation of a novel
J.L. Pedraz, Long-term expression of erythropoietin from myoblasts immobilized in phytosome-loaded chitosan microsphere system for curcumin delivery, Int. J.
biocompatible and neovascularized microcapsules, Mol. Ther. 12 (2005) 283–289. Pharm. 448 (2013) 168–174.
[72] A.Z. Chen, M.Y. Chen, S.B. Wang, X.N. Huang, Y.G. Liu, Z.X. Chen, Poly(L-histidine)– [101] X.Y. Li, L.J. Jin, J.E. Uzonna, S.Y. Li, J.J. Liu, H.Q. Li, Y.N. Lu, Y.H. Zhen, Y.P. Xu,
chitosan/alginate complex microcapsule as a novel drug delivery agent, J. Appl. Chitosan–alginate microcapsules for oral delivery of egg yolk immunoglobulin
Polym. Sci. 124 (2012) 3728–3736. (IgY): in vivo evaluation in a pig model of enteric colibacillosis, Vet. Immunol.
[73] K. Shimokawa, K. Saegusa, Y. Wada, F. Ishii, Physicochemical properties and con- Immunopathol. 129 (2009) 132–136.
trolled drug release of microcapsules prepared by simple coacervation, Colloids [102] Y. Zhang, W. Wei, P. Lv, L. Wang, G. Ma, Preparation and evaluation of alginate–
Surf. B: Biointerfaces 104 (2013) 1–4. chitosan microspheres for oral delivery of insulin, Eur. J. Pharm. Biopharm. 77
[74] S. Harada, S. Ehara, K. Ishii, H. Yamazaki, S. Matsuyama, T. Sato, S. Oikawa, T. Kamiya, (2011) 11–19.
K. Arakawa, W. Yokota, K. Sera, J. Ito, Targeted delivery of chemotherapeutic agents [103] D. Tahtat, M. Mahlous, S. Benamer, A.N. Khodja, H. Oussedik-Oumehdi, F.
using improved radiosensitive liquid core microcapsules and assessment of their Laraba-Djebari, Oral delivery of insulin from alginate/chitosan crosslinked by
antitumor effect, Int. J. Radiat. Oncol. Biol. Phys. 75 (2009) 455–462. glutaraldehyde, Int. J. Biol. Macromol. 58 (2013) 160–168.
[75] A.W. Scott, B.M. Tyler, B.C. Masi, U.M. Upadhyay, Y.R. Patta, R. Grossman, L. [104] C. Santivarangkna, U. Kulozik, P. Foerst, Alternative drying processes for the
Basaldella, R.S. Langer, H. Brem, M.J. Cima, Intracranial microcapsule drug delivery industrial preservation of lactic acid starter cultures, Biotechnol. Prog. 23 (2007)
device for the treatment of an experimental gliosarcoma model, Biomaterials 32 302–315.
(2011) 2532–2539. [105] Z. Fang, B. Bhandari, Encapsulation techniques for food ingredient systems, in: B.
[76] S.J. Park, Y.M. Lee, S.K. Hong, Release behaviors of porous poly(butylene Bhandari, Y.H. Roos (Eds.), Food Materials Science and Engineering, Wiley-Blackwell,
succinate)/poly(ɛ-caprolactone) microcapsules containing indomethacin, Colloids Oxford, 2012, pp. 320–344.
Surf. B: Biointerfaces 47 (2006) 211–215. [106] H. Ichikawa, Y. Fukumori, C.M. Adeyeye, Design of prolonged-release microcap-
[77] H. Hamishehkar, J. Emami, A.R. Najafabadi, K. Gilani, M. Minaiyan, K. Hassanzadeh, sules containing diclofenac sodium for oral suspensions and their preparation by
H. Mahdavi, M. Koohsoltani, A. Nokhodchi, Pharmacokinetics and pharmacody- the Wurster process, Int. J. Pharm. 156 (1997) 39–48.
namics of controlled release insulin loaded PLGA microcapsules using dry powder [107] S. Santoyo, E.G. de Jalón, P. Ygartua, M.J. Renedo, M.J. Blanco-Príeto, Optimization of
inhaler in diabetic rats, Biopharm. Drug Dispos. 31 (2010) 189–201. topical cidofovir penetration using microcapsules, Int. J. Pharm. 242 (2002)
[78] N.V.N. Jyothi, P.M. Prasanna, S.N. Sakarkar, K.S. Prabha, P.S. Ramaiah, G.Y. Srawan, 107–113.
Microencapsulation techniques, factors influencing encapsulation efficiency, [108] M. Hrynyk, M. Martins-Green, A.E. Barron, R.J. Neufeld, Sustained prolonged topical
J. Microencapsul. 27 (2010) 187–197. delivery of bioactive human insulin for potential treatment of cutaneous wounds,
[79] N. Ngwuluka, Application of in situ polymerization for design and development of Int. J. Pharm. 398 (2010) 146–154.
oral drug delivery systems, AAPS PharmSciTech 11 (2010) 1603–1611. [109] Y. Zhang, Z. Zhi, X. Li, J. Gao, Y. Song, Carboxylated mesoporous carbon microparti-
[80] M. Morishita, A.M. Lowman, K. Takayama, T. Nagai, N.A. Peppas, Elucidation of the cles as new approach to improve the oral bioavailability of poorly water-soluble
mechanism of incorporation of insulin in controlled release systems based on carvedilol, Int. J. Pharm. 454 (2013) 403–411.
complexation polymers, J. Control. Release 81 (2002) 25–32. [110] A. Madene, M. Jacquot, J. Scher, S. Desobry, Flavour encapsulation and controlled
[81] M. Morishita, T. Goto, N.A. Peppas, J.I. Joseph, M.C. Torjman, C. Munsick, K. release — a review, Int. J. Food Sci. Technol. 41 (2006) 1–21.
Nakamura, T. Yamagata, K. Takayama, A.M. Lowman, Mucosal insulin delivery sys- [111] G.A. Reineccius, Flavor encapsulation, Food Rev. Int. 5 (1989) 147–176.
tems based on complexation polymer hydrogels: effect of particle size on insulin [112] P. Giunchedi, U. Conte, Spray-drying as a preparation method of microparticulate
enteral absorption, J. Control. Release 97 (2004) 115–124. drug delivery systems: an overview, S.T.P. Pharma. Sci. 5 (1995) 276–290.
[82] M. Morishita, T. Goto, K. Nakamura, A.M. Lowman, K. Takayama, N.A. Peppas, Novel [113] D.X. Li, Y.K. Oh, S.J. Lim, J.O. Kim, H.J. Yang, J.H. Sung, C.S. Yong, H.G. Choi, Novel
oral insulin delivery systems based on complexation polymer hydrogels: single gelatin microcapsule with bioavailability enhancement of ibuprofen using
and multiple administration studies in type 1 and 2 diabetic rats, J. Control. Release spray-drying technique, Int. J. Pharm. 355 (2008) 277–284.
110 (2006) 587–594. [114] R.P. Raffin, L.M. Colomé, C.R.D. Hoffmeister, P. Colombo, A. Rossi, F. Sonvico, L.M.
[83] H.G. Bungenberg de Jong, H.R. Kruyt, Coacervation (partial miscibility in colloid Colomé, C.C. Natalini, A.R. Pohlmann, T.D. Costa, S.S. Guterres, Pharmacokinetics
systems), Proc. K. Ned. Akad. Wet. 32 (1929) 849–856. evaluation of soft agglomerates for prompt delivery of enteric pantoprazole-loaded
[84] H.G. Bungenberg de Jong, Crystallization–coacervation–flocculation, in: H.R. Kruyt microparticles, Eur. J. Pharm. Biopharm. 74 (2010) 275–280.
(Ed.), Colloid Sci.Elsevier, Amsterdam, 1949, pp. 232–258. [115] S. Bajaj, D. Singla, N. Sakhuja, Stability testing of pharmaceutical products, J. Appl.
[85] R. Dubey, T.C. Shami, K.U. Bhasker Rao, Microencapsulation technology and Pharm. Sci. 02 (2012) 129–138.
applications, Def. Sci. J. 59 (2009) 82–95. [116] In: S. Yoshioka, V.J. Stella (Eds.), Stability of Drugs and Dosage Forms, Kluwer
[86] C. Tomaro–Duchesneau, S. Saha, M. Malhotra, I. Kahouli, S. Prakash, Microencapsu- Academic, New York, 2000.
lation for the therapeutic delivery of drugs, live mammalian and bacterial cells, and [117] A.D. Deshpande, K.G. Baheti, N.R. Chatterjee, Degradation of β-lactam antibiotics,
other biopharmaceutics: current status and future directions, J. Pharm. 2013 Curr. Sci. 87 (2004) 1684–1695.
(2013) 103527. [118] J.R. Delanghe, M.R. Langlois, M.L. de Buyzere, M.A. Torck, Vitamin C deficiency and
[87] J.S. Patil, M.V. Kamalapur, S.C. Marapur, D.V. Kadam, Ionotropic gelation and poly- scurvy are not only a dietary problem but are codetermined by the haptoglobin
electrolyte complexation: the novel techniques to design hydrogel particulate polymorphism, Clin. Chem. 53 (2007) 1397–1400.
sustained, modulated drug delivery system: a review, Dig. J. Nanomater. Biostruct. [119] W. Wang, Instability, stabilization, and formulation of liquid protein pharmaceuti-
5 (2010) 241–248. cals, Int. J. Pharm. 185 (1999) 129–188.
[88] P. Patil, D. Chavanke, M. Wagh, A review on ionotropic gelation method: novel [120] T. Handa, S. Singh, I.P. Singh, Characterization of a new degradation product of ni-
approach for controlled gastroretentive gelispheres, Int. J. Pharm. Pharm. Sci. 4 fedipine catalyzed by atenolol: a typical case of alteration of degradation pathway
(2012) 27–32. of one drug by another, J. Pharm. Biomed. Anal. 89 (2014) 6–17.
[89] Wurster, D.E., Method of applying coatings to edible tablets or the like, US Patent [121] M. Allwood, A. Stanley, P. Wright, The Cytotoxics Handbook, Radcliffe Medical
2648609 (1953). Press Ltd., UK, 2002, p. 333, (361).
[90] C. Peniche, W. Argűelles-Monal, H. Peniche, N. Acosta, Chitosan: an attractive [122] G. Vilar, J. Tulla-Puche, F. Albericio, Polymers and drug delivery systems, Curr. Drug
biocompatible polymer for microencapsulation, Macromol. Biosci. 3 (2003) Deliv. 9 (2012) 367–394.
511–520. [123] K.E. Uhrich, S.M. Cannizzaro, R.S. Langer, K.M. Shakesheff, Polymeric systems for
[91] S.K. Ghosh, Functional coatings and microencapsulation: a general perspective, in: controlled drug release, Chem. Rev. 99 (1999) 3181–3198.
S.K. Ghosh (Ed.), Functional Coatings: by Polymer Microencapsulation, Wiley-VCH [124] H. Maeda, SMANCS and polymer-conjugated macromolecular drugs: advantages in
Verlag GmbH & Co. KGaA, Weinheim, 2006, pp. 1–28. cancer chemotherapy, Adv. Drug Deliv. Rev. 6 (1991) 181–202.
[92] Y. Yeo, N. Baek, K. Park, Microencapsulation methods for delivery of protein drugs, [125] H. Maeda, T. Konno, Metamorphosis of neocarzinostatin to SMANCS: chemistry,
Biotechnol. Bioprocess Eng. 6 (2001) 213–230. biology, pharmacology, and clinical effect of the first prototype anticancer polymer
[93] P. Couvreur, G. Barrat, E. Fattal, P. Legrand, C. Vauthier, Nanocapsule technology, therapeutic, in: H. Maeda, K. Edo, N. Ishida (Eds.), Neocarzinostatin: The Past,
Crit. Rev. Ther. Drug Carrier Syst. 19 (2002) 99–134. Present, and Future of an Anticancer Drug, Springer Verlag, Tokyo, 1997, pp. 227–267.
[94] W. He, X. Gu, S. Liu, Surfactant-free one-step synthesis of dual-functional polyurea [126] M.J. Vicent, R. Duncan, Polymer conjugates: nanosized medicines for treating
microcapsules: contact infection control and drug delivery, Adv. Funct. Mater. 22 cancer, Trends Biotechnol. 24 (2006) 39–47.
(2012) 4023–4031. [127] Y.E. Kurtoglu, M.K. Mishra, S. Kannan, R.M. Kannan, Drug release characteristics of
[95] N. Kamei, M. Morishita, H. Chiba, N.J. Kavimandan, N.A. Peppas, K. Takayama, PAMAM dendrimer–drug conjugates with different linkers, Int. J. Pharm. 384
Complexation hydrogels for intestinal delivery of interferon β and calcitonin, (2010) 189–194.
J. Control. Release 134 (2009) 98–102. [128] L. Liang, S.W. Lin, W. Dai, J.K. Lu, T.Y. Yang, Y. Xiang, Y. Zhang, R.T. Li, Q. Zhang,
[96] J.M. Ruiz, B. Tissier, J.P. Benoit, Microencapsulation of peptide: a study of the phase Novel cathepsin B-sensitive paclitaxel conjugate: higher water solubility, better
separation of PLGA copolymers 50/50 by silicone oil, Int. J. Pharm. 49 (1989) 69–77. efficacy and lower toxicity, J. Control. Release 160 (2012) 618–629.
[97] S. Takada, Y. Uda, H. Toguchi, Y. Ogawa, Application of a spray drying technique in [129] I.J.C. Castellanos, G. Cruz, R. Crespo, K. Griebenow, Encapsulation-induced aggrega-
the production of TRH-containing injectable sustained-release microparticles of tion and loss in activity of g-chymotrypsin and their prevention, J. Control. Release
biodegradable polymers, PDA J. Pharm. Sci. Technol. 49 (1995) 180–184. 81 (2002) 307–319.
[98] P.F. Builders, O.O. Kunle, L.C. Okpaku, M.I. Builders, A.A. Attama, M.U. Adikwu, [130] K.K. Kim, D.W. Pack, Microspheres for drug delivery, in: M. Ferrari, A.P. Lee, L.J. Lee
Preparation and evaluation of mucinated sodium alginate microparticles for oral (Eds.), BioMEMS and Biomedical Nanotechnology, vol. I Biological and Biomedical
delivery of insulin, Eur. J. Pharm. Biopharm. 70 (2008) 777–783. Nanotechnology, Springer Science + Business Media LLC, New York, 2006,
[99] J. Quan, S.M. Kim, C.H. Pan, D. Chung, Characterization of fucoxanthin-loaded pp. 19–50.
microspheres composed of cetyl palmitate-based solid lipid core and fish gela- [131] D.A. Parkins, U.T. Lashmar, The formulation of biopharmaceutical products, Pharm.
tin–gum arabic coacervate shell, Food Res. Int. 50 (2013) 31–37. Sci. Technol. Today 3 (2000) 129–137.
P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45 43

[132] C. Pérez, I.J. Castellanos, H.R. Constantino, W. Al-Azzam, K. Griebenow, Recent [162] C.W.M. Yuen, J. Yip, L. Liu, K. Cheuk, C.W. Kan, H.C. Cheung, S.Y. Cheng, Chitosan
trends in the stabilization of proteins upon encapsulation in and release from microcapsules loaded with either miconazole nitrate or clotrimazole, prepared
bioerodible polymers, J. Pharm. Pharmacol. 54 (2002) 301–313. via emulsion technique, Carbohydr. Polym. 89 (2012) 795–801.
[133] I.J.C. Castellanos, R. Crespo, K. Griebenow, Poly(ethylene glycol) as stabilizer and [163] E. Yenilmez, E. Başaran, Y. Yazan, Release characteristics of vitamin E incorporated
emulsifying agent: a novel stabilization approach preventing aggregation and chitosan microspheres and in vitro–in vivo evaluation for topical application,
inactivation of proteins upon encapsulation in bioerodible polyester microspheres, Carbohydr. Polym. 84 (2011) 807–811.
J. Control. Release 88 (2003) 135–145. [164] R. Bhati, R.K. Nagrajan, A detail review on oral mucosa of drug delivery system, Int.
[134] E.S. Lee, K.H. Park, I.S. Park, K. Na, Glycol chitosan as a stabilizer for protein encap- J. Pharm. Sci. Res. 3 (2012) 659–681.
sulated into poly(lactide-co-glycolide) microparticle, Int. J. Pharm. 338 (2007) [165] C. Marriott, N.P. Gregory, Mucus physiology and pathology, in: V. Lenaerts, R. Gurny
310–316. (Eds.), Bioadhesive Drug Delivery Systems, CRC Press, Boca Raton, 1990, pp. 1–24.
[135] X.Y. Li, X.G. Chenb, Z.W. Suna, H.J. Parkc, D.S. Cha, Preparation of alginate/ [166] V.V. Khutoryanskiy, Advances in mucoadhesion and mucoadhesive polymers,
chitosan/carboxy methyl chitosan complex microcapsules and application in Macromol. Biosci. 11 (2011) 748–764.
Lactobacillus casei ATCC 393, Carbohydr. Polym. 83 (2011) 1479–1485. [167] I.A. Sogias, A.C. Williams, V.V. Khutoryanskiy, Why is chitosan mucoadhesive?
[136] S. Arora, R.D. Budhiraja, Chitosan–alginate microcapsules of amoxicillin for gastric Biomacromolecules 9 (2008) 1837–1842.
stability and mucoadhesion, J. Adv. Pharm. Technol. Res. 3 (2012) 68–74. [168] H.L. Lueβen, C.M. Lehr, C.O. Rentel, A.B.J. Noach, A.G. de Boer, J.C. Verhoef, H.E.
[137] H.S. Akre, D.R. Mundhada, S. Bhaskaran, S. Asghar, G.S. Gandhi, Dry suspension Junginger, Bioadhesive polymers for the peroral delivery of peptide drugs,
formulation of taste masked antibiotic drug for pediatric use, JAPS 02 (2012) J. Control. Release 29 (1994) 329–338.
166–171. [169] J.R. Robinson, G.M. Mlynek, Bioadhesive and phase-change polymers for ocular
[138] K. Prakash, P.N. Raju, K.K. Shanta, M.N. Lakshmi, Preparation and characterization drug delivery, Adv. Drug Deliv. Rev. 16 (1995) 45–50.
of lamivudine microcapsules using various cellulose polymers, Trop. J. Pharm. [170] P. He, S.S. Davis, L. Illum, In vitro evaluation of the mucoadhesive properties of
Res. 6 (2007) 841–847. chitosan microspheres, Int. J. Pharm. 166 (1998) 75–88.
[139] N.R. Padala, K. Prakash, C.S. Reddy Bonepally, B. Krishnaveni, K. Shantakumari, N.M. [171] J. Shimoda, H. Onishi, Y. Machida, Bioadhesive characteristics of chitosan micro-
Lakshmi, Stavdine loaded microcapsules using various cellulose polymers: prepa- spheres to the mucosa of rat small intestine, Drug Dev. Ind. Pharm. 27 (2001)
ration and in-vitro evaluation, Int. J. Pharm. Sci. Res. 2 (2009) 551–556. 567–576.
[140] M.V. Bule, R.S. Singhal, J.F. Kennedy, Microencapsulation of ubiquinone-10 in [172] E.E. Hassan, J.M. Gallo, A simple rheological method for the in vitro assessment of
carbohydrate matrices for improved stability, Carbohydr. Polym. 82 (2010) mucin-polymer bioadhesive bond strength, Pharm. Res. 7 (1990) 491–495.
1290–1296. [173] V.R. Sinha, A.K. Singla, S. Wadhawan, R. Kaushik, R. Kumria, K. Bansal, S. Dhawan,
[141] C.W. Hsieh, W.Ch. Lu, W.C. Hsieh, Y.P. Huang, C.H. Lai, W.C. Ko, Improvement of the Chitosan microspheres as a potential carrier for drugs, Int. J. Pharm. 274 (2004)
stability of nattokinase using γ-polyglutamic acid as a coating material for micro- 1–33.
encapsulation, LWT Food Sci. Technol. 42 (2009) 144–149. [174] S. Dhawan, A.K. Singla, V.R. Sinha, Evaluation of mucoadhesive properties of chito-
[142] R.A. Siegel, M.J. Rathbone, Overview of controlled release mechanisms, in: J. san microspheres prepared by different methods, AAPS PharmSciTech 5 (2004)
Siepmann, R.A. Siegel, M.J. Rathbone (Eds.), Fundamentals and Applications 1–7.
of Controlled Release Drug Delivery, 1st edition, Springer, New York, 2012, [175] A.S. Pedro, E. Cabral-Albuquerque, D. Ferreira, B. Sarmento, Chitosan: an option for
pp. 19–46. development of essential oil delivery systems for oral cavity care? Carbohydr.
[143] K. Kooiman, M.R. Böhmer, M. Emmer, H.J. Vos, C. Chlon, W.T. Shi, C.S. Hall, S.H.P.M. Polym. 76 (2009) 501–508.
de Winter, K. Schroën, M. Versluis, N. de Jong, A. van Wamel, Oil-filled polymer mi- [176] I. Bravo-Osuna, C. Vauthier, A. Farabollini, G.F. Palmieri, G. Ponchel, Mucoadhesion
crocapsules for ultrasound-mediated delivery of lipophilic drugs, J. Control. Release mechanism of chitosan and thiolated chitosan–poly(isobutyl cyanoacrylate) core–
133 (2009) 109–118. shell nanoparticles, Biomaterials 28 (2007) 2233–2243.
[144] J.C. Jeong, J. Lee, K. Cho, Effects of crystalline microstructure on drug release behav- [177] L. Yin, J. Ding, C. He, L. Cui, C. Tang, C. Yin, Drug permeability and mucoadhesion
ior of poly(ε-caprolactone) microspheres, J. Control. Release 92 (2003) 249–258. properties of thiolated trimethyl chitosan nanoparticles in oral insulin delivery,
[145] P. Borgquist, A. Körner, L. Piculell, A. Larsson, A. Axelsson, A model for the drug re- Biomaterials 30 (2009) 5691–5700.
lease from a polymer matrix tablet—effects of swelling and dissolution, J. Control. [178] E. Hagesaether, M. Hiorth, S.A. Sande, Mucoadhesion and drug permeability of free
Release 113 (2006) 216–225. mixed films of pectin and chitosan: an in vitro and ex vivo study, Eur. J. Pharm.
[146] G. Fundueanu, M. Constantin, E. Esposito, R. Cortesi, C. Nastruzzi, E. Menegatti, Cel- Biopharm. 71 (2009) 325–331.
lulose acetate butyrate microcapsules containing dextran ion-exchange resins as [179] F.A. Moghaddam, F. Atyabi, R. Dinarvand, Preparation and in vitro evaluation of
self-propelled drug release system, Biomaterials 26 (2005) 4337–4347. mucoadhesion and permeation enhancement of thiolated chitosan-pHEMA core–
[147] K.S. Kim, S.J. Park, Characterization and release behaviors of porous PCL/Eudragit shell nanoparticles, Nanomedicine 5 (2009) 208–215.
RS microcapsules containing tulobuterol, Colloids Surf. B: Biointerfaces 76 (2010) [180] B.E. Benediktsdóttir, Ó. Baldursson, M. Másson, Challenges in evaluation of chitosan
404–409. and trimethylated chitosan (TMC) as mucosal permeation enhancers: from syn-
[148] R.V. Kulkarni, B.S. Mangond, S. Mutalik, B. Sa, Interpenetrating polymer network thesis to in vitro application, J. Control. Release 173 (2014) 18–31.
microcapsules of gellan gum and egg albumin entrapped with diltiazem–resin com- [181] S.M. Schneider, K. Hess, T. Gosselin, Interventions to promote adherence with oral
plex for controlled release application, Carbohydr. Polym. 83 (2011) 1001–1007. agents, Semin. Oncol. Nurs. 27 (2011) 133–141.
[149] S. Fredenberg, M. Wahlgren, M. Reslow, A. Axelsson, The mechanisms of drug [182] J.C. Kim, H.Y. Lee, M.H. Kim, H.J. Lee, H.Y. Kang, S.M. Kim, Preparation and charac-
release in poly(lactic-co-glycolic acid)-based drug delivery systems—A review, terisation of chitosan/gelation microcapsules containing triclosan, Colloids Surf.
Int. J. Pharm. 415 (2011) 34–52. B: Biointerfaces 52 (2006) 52–56.
[150] A. Zhu, F. Li, L. Ji, Poly(lactic acid)/N-maleoylchitosan core–shell capsules: prepara- [183] K.S. Huang, K. Lu, C.S. Yeh, S.R. Chung, C.H. Lin, C.H. Yang, Y.S. Dong, Microfluidic
tion and drug release properties, Colloids Surf. B: Biointerfaces 91 (2012) 162–167. controlling monodisperse microdroplet for 5-fluorouracil loaded genipin–gelatin
[151] H. Umer, H. Nigam, A.M. Tamboli, M.S.M. Nainar, Microencapsulation: process, microcapsules, J. Control. Release 137 (2009) 15–19.
techniques and applications, Int. J. Res. Pharm. Biomed. Anal. Sci. 2 (2011) [184] B. Jamilah, K.G. Harvinder, Properties of gelatins from skins of fish-black tilapia
474–481. (Oreochromis mossambicus) and red tilapia (Oreochromis nilotica), Food Chem. 77
[152] Q.X. Wu, S.J. Yao, Novel NaCS–CS–PPS microcapsules as a potential enzyme- (2002) 81–84.
triggered release carrier for highly-loading 5-ASA, Colloids Surf. B: Biointerfaces [185] P.M. Gilsenan, S.B. Ross-Murphy, Rheological characterization of gelatins from
109 (2013) 147–153. mammalian and marine sources, Food Hydrocolloids 14 (2000) 191–195.
[153] R.K. Verma, S. Arora, S. Garg, Osmotic pumps in drug delivery, Crit. Rev. Ther. Drug [186] I.S. Arvanitoyanni, Protein-Based Films and Coatings, CRC Press, 2002.
Carrier Syst. 21 (2004) 477–520. [187] R. Wakode, A. Baja, Gelatin microspheres for topical delivery of vitamin A
[154] S. Herrlich, S. Spieth, S. Messner, R. Zengerle, Osmotic micropumps for drug deliv- palmitate, NSHM J. Pharm. Healthc. Manag. 02 (2011) 61–65.
ery, Adv. Drug Deliv. Rev. 64 (2012) 1617–1627. [188] P.L. Lam, S.H.L. Kok, Y.W. Ho, R.S.M. Wong, G.Y.M. Cheng, C.H. Cheng, K.H. Lam, R.
[155] W. Ryu, Z. Huang, F.B. Prinz, S.B. Goodman, R. Fasching, Biodegradable micro- Gambari, K.K.H. Lee, C.H. Chui, A novel green gelatin-agar microencapsulation
osmotic pump for long-term and controlled release of basic fibroblast growth system with P. urinaria as an improved anti-A. niger model, Carbohydr. Polym. 92
factor, J. Control. Release 124 (2007) 98–105. (2013) 877–880.
[156] I. El-Gibaly, Development and in vitro evaluation of novel floating chitosan micro- [189] C.M. Ofner III, H. Schott, Swelling studies of gelatin I: gelatin without additives,
capsules for oral use: comparison with non-floating chitosan microcapsules, Int. J. J. Pharm. Sci. 75 (1986) 790–796.
Pharm. 249 (2002) 7–21. [190] J.R. Gillmor, R.W. Connelly, R.H. Colby, J.S. Tan, Effect of sodium poly(styrene
[157] A.A. Sunil, N.M. Nadagouda, M.A. Tejraj, Recent advances on chitosan-based micro- sulfonate) on thermoreversible gelation of gelatin, J. Polym. Sci. B Polym. Phys.
and nanoparticles in drug delivery, J. Control. Release 100 (2004) 5–28. 37 (1999) 2287–2295.
[158] W. Tiyaboonchai, Chitosan nanoparticles: a promising system for drug delivery, [191] J.H. Choi, V. Loftness, Investigation of human body skin temperatures as a bio-signal
Naresuan Univ. J. 11 (2003) 51–66. to indicate overall thermal sensations, Build. Environ. 58 (2012) 258–269.
[159] R.J. Soane, M. Frier, A.C. Perkins, N.S. Jones, S.S. Davis, L. Illum, Evaluation of the [192] G. Thakur, A. Mitra, A. Basak, D. Sheet, Characterization and scanning electron
clearance characteristics of bioadhesive systems in humans, Int. J. Pharm. 178 microscopic investigation of crosslinked freeze dried gelatin matrices for study of
(1999) 55–65. drug diffusivity and release kinetics, Micron 43 (2012) 311–320.
[160] S.Y. Cheng, M.C.W. Yuen, P.L. Lam, R. Gambari, R.S.M. Wong, G.Y.M. Cheng, P.B.S. [193] H. Yasuda, C. Lamaze, L.D. Ikenberry, Permeability of solutes through hydrated
Lai, S.W. Tong, K.W. Chan, F.Y. Lau, S.H.L. Kok, K.H. Lam, C.H. Chui, Synthesis, polymer membranes. I: Diffusion of sodium chloride, Makromol. Chem. 118
characterization and preliminary analysis of in vivo biological activity of (1968) 19–35.
chitosan/celecoxib microcapsules, Bioorg. Med. Chem. Lett. 20 (2010) 4147–4151. [194] P.L. Lam, L. Li, C.W.M. Yuen, R. Gambari, R.S.M. Wong, C.H. Chui, K.H. Lam, Effects of
[161] H. Lee, C. Jeong, K. Ghafoor, S. Cho, J. Park, Oral delivery of insulin using chitosan multiple washing on cotton fabrics containing berberine microcapsules with
capsules cross-linked with phytic acid, Biomed. Mater. Eng. 21 (2011) 25–36. anti-Staphylococcus aureus activity, J. Microencapsul. 30 (2013) 143–150.
44 P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45

[195] P. Sriamornsak, J. Nunthanid, M. Luangtana-Anan, Y. Weerapol, S. Puttipipatkhachorn, [225] S.J. Park, K.S. Kim, Effect of oxygen plasma treatment on the release behaviors of
Alginate-based pellets prepared by extrusion/spheronization: effect of the amount poly(ɛ-caprolactone) microcapsules containing tocopherol, Colloids Surf. B:
and type of sodium alginate and calcium salts, Eur. J. Pharm. Biopharm. 69 (2008) Biointerfaces 43 (2005) 138–142.
274–284. [226] M.H. Perez, C. Zinutti, A. Lamprecht, N. Ubrich, A. Astier, M. Hoffman, R. Bodmeier,
[196] K. Pal, A.T. Paulson, D. Rousseau, Biopolymers in controlled-delivery release P. Maincent, The preparation and evaluation of poly( -caprolactone) microparticles
systems, in: S. Ebnesajjad (Ed.), Handbook of Biopolymers and Biodegradable containing both a lipophilic and a hydrophilic drug, J. Control. Release 65 (2000)
Plastics: Properties, Processing and Applications, Elsevier Inc., USA, 2013, 429–438.
pp. 329–364. [227] M.A. Benoit, B. Baras, J. Gillard, Preparation and characterization of protein-loaded
[197] R.C. Rowe, P.J. Shesky, P.J. Weoller, Hand Book of Pharmaceutical Excipients, Fourth poly(o-caprolactone) microparticles for oral vaccine delivery, Int. J. Pharm. 184
ed. Pharm Press, London, UK, 2003. (1999) 73–84.
[198] L. Liu, F. Wu, X.J. Ju, R. Xie, W. Wang, C.H. Niu, L.Y. Chu, Preparation of monodisperse [228] F.M. Barboza, W.M. Machado, L.R.O. Junior, S.F. Zawadzki, D. Fernandes, P.V. Farago,
calcium alginate microcapsules via internal gelation in microfluidic-generated PCL/PHBV microparticles as innovative carriers for oral controlled release of
double emulsions, J. Colloid Interface Sci. 404 (2013) 85–90. manidipine dihydrochloride, Sci. World J. (2014)(http://www.hindawi.com/
[199] R. Rastogi, Y. Sultana, M. Aqil, A. Ali, S. Kumar, K. Chuttani, A.K. Mishra, Alginate journals/tswj/aip/268107/) (in press).
microspheres of isoniazid for oral sustained drug delivery, Int. J. Pharm. 334 [229] N. Yuksel, M. Baykara, H. Shirinzade, S. Suzen, Investigation of triacetin effect on
(2007) 71–77. indomethacin release from poly(methyl methacrylate) microspheres: evaluation
[200] R.A.H. Ishak, G.A.S. Awad, N.D. Mortada, S.A.K. Nour, Preparation, in vitro and in vivo of interactions using FT-IR and NMR spectroscopies, Int. J. Pharm. 404 (2011)
evaluation of stomach-specific metronidazole-loaded alginate beads as local 102–109.
anti-Helicobacter pylori therapy, J. Control. Release 119 (2007) 207–214. [230] L. Solhi, M. Atai, A. Nodehi, M. Imani, A novel dentin bonding system containing
[201] R.N. Sandhya, K.M. Teja, V. Saikishore, Design and development of sweet potato poly(methacrylic acid) grafted nanoclay: synthesis, characterization and proper-
starch blended sodium alginate mucoadhesive microcapsules of glipizide, Res. J. ties, Dent. Mater. 28 (2012) 1041–1050.
Pharm. Dosage Forms Technol. 4 (2012) 119–123. [231] Q. Xu, W. Huang, L. Jiang, Z. Lei, X. Li, H. Deng, KGM and PMAA based pH-sensitive
[202] N. Hoenich, Cellulose for medical applications: past, present and future, Bioresources 1 interpenetrating polymer network hydrogel for controlled drug release,
(2006) 270–280. Carbohydr. Polym. 97 (2013) 565–570.
[203] S.K. Prajapati, P. Tripathi, U. Ubaidulla, V. Anand, Design and development of [232] M. Andersson Trojer, A. Wendel, K. Holmberg, M. Nydén, The effect of pH on
gliclazide mucoadhesive microcapsules: in vitro and in vivo evaluation, AAPS charge, swelling and desorption of the dispersant poly(methacrylic acid) from
PharmSciTech 9 (2008) 224–230. poly(methyl methacrylate) microcapsules, J. Colloid Interface Sci. 375 (2012)
[204] D. Pal, A.K. Nayak, Development, optimization, and anti-diabetic activity of 213–215.
gliclazide-loaded alginate–methyl cellulose mucoadhesive microcapsules, AAPS [233] J. Kovacs-Nolan, Y. Mine, Microencapsulation for the gastric passage and controlled
PharmSciTech 12 (2011) 1431–1441. intestinal release of immunoglobulin Y, J. Immunol. Methods 296 (2005) 199–209.
[205] G. Murtaza, M. Ahmad, N. Akhtar, Niowaiver study of oral tabletted ethylcellulose [234] G. Año, A. Esquisabel, M. Pastor, A. Talavera, B. Cedré, S. Fernández, S. Sifontes, Y.
microcapsules of a BCS class I drug, Bull. Chem. Soc. Ethiop. 23 (2009) 175–186. Aranguren, G. Falero, L. García, R.L. Solís, J.L. Pedraz, A new oral vaccine candidate
[206] L. Hu, W. Liu, L. Li, J. Zhao, X. Yang, Preparation and in vitro, in vivo evaluation of based on the microencapsulation by spray-drying of inactivated Vibrio cholerae,
clarithromycin microcapsules, JBCP 002 (2011) 1–9. Vaccine 29 (2011) 5758–5764.
[207] G. Giandalia, V. De Caro, L. Cordone, L.I. Giannola, Trehalose–hydroxyethylcellulose [235] E. Dickinson, D.J. McClements, Water-in-oil-in-water multiple emulsion, in: E.
microspheres containing vancomycin for topical drug delivery, Eur. J. Pharm. Dickinson, D.J. McClements (Eds.), Advances in Food Colloids, Blackie Academic
Biopharm. 52 (2001) 83–89. & Professional, London, 1995, pp. 280–300.
[208] N. Wakiyama, K. Juni, M. Nakano, Preparation and evaluation in vitro and in vivo of [236] C. Lobato-Calleros, M.T. Recillas-Mota, T. Espinosa-Solares, J. Alvarez-Ramirez, E.J.
polylactic acid microspheres containing dibucaine, Chem. Pharm. Bull. 30 (1982) Vernon-Carter, Microstructural and rheological properties of low-fat stirred yoghurts
3719–3727. made with skim milk and multiple emulsions, J. Texture Stud. 40 (2009) 657–675.
[209] R.A. Jain, The manufacturing techniques of various drug loaded biodegradable poly [237] I. Danielsson, B. Lindman, The definition of a microemulsion, Colloids Surf. 3
(lactide-co-glycolide) (PLGA) devices, Biomaterials 21 (2000) 2475–2490. (1981) 391–392.
[210] J.W. Rhim, Preparation and characterization of vacuum sputter silver coated PLA [238] T. Kawakatsu, G. Trägårdh, C. Trägårdh, Production of W/O/W emulsions and
film, LWT Food Sci. Technol. 54 (2013) 477–484. S/O/W pectin microcapsules by microchannel emulsification, Colloids Surf. A:
[211] H. Deng, Z. Lei, Preparation and characterization of hollow Fe3O4/SiO2@PEG–PLA Physicochem. Eng. Aspects 189 (2001) 257–264.
nanoparticles for drug delivery, Compos. Part B Eng. 54 (2013) 194–199. [239] T. Foster, K.D. Dorfman, H.T. Davis, Giant biocompatible and biodegradable PEG–
[212] A.N. Ford Versypt, D.W. Pack, R.D. Braatz, Mathematical modeling of drug delivery PMCL vesicles and microcapsules by solvent evaporation from double emulsion
from autocatalytically degradable PLGA microspheres — A review, J. Control. droplets, J. Colloid Interface Sci. 351 (2010) 140–150.
Release 165 (2013) 29–37. [240] P.J. Cai, Y.J. Tang, Y.T. Wang, Y.J. Cao, Fabrication of polystyrene hollow spheres in
[213] J.M. Anderson, M.S. Shive, Biodegradation and biocompatibility of PLA and PLGA W/O/W multiple emulsions, Mater. Chem. Phys. 124 (2010) 10–12.
microspheres, Adv. Drug Deliv. Rev. 28 (1997) 5–24. [241] R. Liu, S.S. Huang, Y.H. Wan, G.H. Ma, Z.G. Su, Preparation of insulin-loaded
[214] X.Y. Ma, G.M. Pan, Z. Lu, J.S. Hu, J.Z. Bei, J.H. Jia, S.G. Wang, Preliminary study of oral PLA/PLGA microcapsules by a novel membrane emulsification method and its
polylactide microcapsulated insulin in vitro and in vivo, Diabetes Obes. Metab. 2 release in vitro, Colloids Surf. B: Biointerfaces 51 (2006) 30–38.
(2000) 243–250. [242] A.J. Khopade, K.S. Nandakumar, N.K. Jainb, Lectin-functionalized multiple
[215] T.K. Mandal, L.A. Bostanian, R.A. Graves, S.R. Chapman, I. Womack, Development of emulsions for improved cancer therapy, J. Drug Target. 6 (1998) 285–292.
biodegradable microcapsules as carrier for oral controlled delivery of amifostine, [243] S.Y. Tang, M. Sivakumar, A.M.H. Ng, P. Shridharan, Anti-inflammatory and analgesic
Drug Dev. Ind. Pharm. 28 (2002) 339–344. activity of novel oral aspirin-loaded nanoemulsion and nano multiple emulsion for-
[216] J. Tian, X. Sun, X. Chen, J. Yu, L. Qu, L. Wang, The formulation and immunisation of mulations generated using ultrasound cavitation, Int. J. Pharm. 430 (2012) 299–306.
oral poly(DL-lactide-co-glycolide)microcapsules containing a plasmid vaccine [244] S. Peltola, P. Saarinen-Savolainen, J. Kiesvaara, T.M. Suhonen, A. Urtti, Microemulsions
against lymphocystis disease virus in Japanese flounder (Paralichthys olivaceus), for topical delivery of estradiol, Int. J. Pharm. 254 (2003) 99–107.
Int. Immunopharmacol. 8 (2008) 900–908. [245] W. Zhu, C. Guo, A. Yu, Y. Gao, F. Cao, G. Zhai, Microemulsion-based hydrogel formu-
[217] E.G. de Jalón, M.J. Blanco-Príeto, P. Ygartua, S. Santoyo, Topical application of lation of penciclovir for topical delivery, Int. J. Pharm. 378 (2009) 152–158.
acyclovir-loaded microparticles: quantification of the drug in porcine skin layers, [246] F. Cournarie, M.P. Savelli, V. Rosilio, F. Bretez, C. Vauthier, J.L. Grossiord, M. Seiller,
J. Control. Release 75 (2001) 191–197. Insulin-loaded W/O/W multiple emulsions: comparison of the performances of
[218] T.M. Don, C.F. King, W.Y. Chiu, C.A. Peng, Preparation and characterization of systems prepared with medium-chain-triglycerides and fish oil, Eur. J. Pharm.
chitosan-g-poly(vinyl alcohol)/poly(vinyl alcohol) blends used for the eval- Biopharm. 58 (2004) 477–482.
uation of blood-contacting compatibility, Carbohydr. Polym. 63 (2006) [247] A. Shahiwala, M.M. Amiji, Enhanced mucosal and systemic immune response with
331–339. squalane oil-containing multiple emulsions upon intranasal and oral administra-
[219] G. Fundueanu, M. Constantin, P. Ascenzi, Poly(vinyl alcohol) microspheres with tion in mice, J. Drug Target. 16 (2008) 302–310.
pH- and thermosensitive properties as temperature-controlled drug delivery, [248] S. Paul, A. Kumar, P. Yedurkar, K. Sawant, Design and development of multiple
Acta Biomater. 6 (2010) 3899–3907. emulsion for enhancement of oral bioavailability of acyclovir, Drug Dev. Ind.
[220] A.R. Bachtsi, C. Kiparissides, Synthesis and release studies of oil-containing Pharm. 39 (2013) 1809–1817.
poly(vinyl alcohol) microcapsules prepared by coacervation, J. Control. [249] F. Tirnaksiz, O. Kalsin, A topical w/o/w multiple emulsions prepared with
Release 38 (1996) 49–58. Tetronic®908 as a hydrophilic surfactant: Formulation, characterization and
[221] S. Kaity, J. Isaac, A. Ghosh, Interpenetrating polymer network of locust bean release study, J. Pharm. Pharm. Sci. 8 (2005) 299–315.
gum-poly (vinyl alcohol) for controlled release drug delivery, Carbohydr. Polym. [250] T. Schmidts, K. Marquardt, P. Schlupp, D. Dobler, F. Heinz, U. Mäder, H. Garn, H.
94 (2013) 456–467. Renz, J. Zeitvogel, T. Werfel, F. Runkel, Development of drug delivery systems for
[222] N. Jain, A. Banik, A. Gupta, Novel interpenetrating polymer network microspheres the dermal application of therapeutic DNAzymes, Int. J. Pharm. 431 (2012) 61–69.
of Lepidium sativum and poly (vinyl alcohol) for the controlled release of simva- [251] G. Orive, R.M. Hernández, A.R. Gascón, R. Calafiore, T.M.S. Chang, P. de Vos, G.
statin, Int. J. Pharm. Pharm. Sci. 5 (2013) 125–130. Hortelano, D. Hunkeler, I. Lacı́k, A.M. Shapiro, J.L. Pedraz, Cell encapsulation:
[223] N. Jain, A. Banik, Novel interpenetrating polymer network mucoadhesive micro- promise and progress, Nat. Med. 9 (2003) 104–107.
spheres of gum ghatti and poly (vinyl alcohol) for the delivery of ranitidine HCl, [252] G. Orive, R.M. Hernández, A.R. Gascón, R. Calafiore, T.M.S. Chang, P. de Vos, G.
Asian J. Pharm. Clin. Res. 6 (2013) 119–123. Hortelano, D. Hunkeler, I. Lacı́k, J.L. Pedraz, History, challenges and perspectives
[224] S.J. Park, Y.J. Yang, H.B. Lee, Effect of acid–base interaction between silica and of cell microencapsulation, Trends Biotechnol. 22 (2004) 87–92.
fragrant oil in the PCL/PEG microcapsules, Colloids Surf. B: Biointerfaces 38 [253] Benke, K.; Bucheler, M.; Michaelis, J.; Pollinger, N.; Rupp, R., Flavor-masked
(2004) 35–40. pharmaceutical compositions, US patent No. 5,695,784, December 9, 1997.
P.L. Lam, R. Gambari / Journal of Controlled Release 178 (2014) 25–45 45

[254] M.L. Jones, C.J. Martoni, M. Parent, S. Prakash, Cholesterol-lowering efficacy of a T.J. Schnitzer, L.B. Trigg, I. Whatmough, A.G. Schmidt, Microemulsion formu-
microencapsulated bile salt hydrolase-active Lactobacillus reuteri NCIMB 30242 lation of cyclosporin (Sandimmun Neoral®) vs Sandimmun®: comparative
yoghurt formulation in hypercholesterolaemic adults, Br. J. Nutr. 107 (2012) safety, tolerability and efficacy in severe active rheumatoid arthritis,
1505–1513. Rheumatology 39 (2000) 156–164.
[255] Ther-Rx Corporation, MICRO-K EXTENCAPS (potassium chloride) capsule, extend- [258] S.K. Jha, S. Dey, R. Karki, Microemulsions—potential carrier for improved drug
ed release MICRO-K 10 EXTENCAPS (potassium chloride) capsule, extended delivery, Asian J. Biomed. Pharm. Sci. 1 (2011) 5–9.
release, U.S. Patent No. 4,259,315, August 1, 2009. [259] E. Kvitnitsky, N. Lerner, Y.E. Shapiro, Tagravit™ microcapsules as controlled drug
[256] Dwyer, M.; Evans, A.M.; Lukas, S., l.H. Pitman, Taste masked pharmaceutical delivery devices and their formulations, in: M.R. Rosen (Ed.), Delivery System
compositions, US patent No. 20,030,064,108, April 3, 2003. Handbook for Personal Care and Cosmetic Products, William Andrew, Inc., Now
[257] D.E. Yocum, S. Allard, S.B. Cohen, P. Emery, R.M. Flipo, J. Goobar, S. York, 2005, pp. 215–258.
Jayawardena, C. Job-Deslandre, R.W. Jubb, K. Krüger, A. Lopes Vaz, B. [260] DS Laboratories, Inc., Product information sheet of Trioxil anti-acne gel,
Manger, E. Mur, H. Nygaard, S.M. Weiner, F. Rainer, M.R. Sack, M.H. Schiff, 2005.

Potrebbero piacerti anche