Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Journal of Pharmaceutical Investigation (2017) 47:287–296 Online ISSN 2093-6214

DOI 10.1007/s40005-017-0320-1 Print ISSN 2093-5552

REVIEW

Mechanisms of drug release from advanced drug formulations


such as polymeric-based drug-delivery systems and lipid
nanoparticles
Gi‑Ho Son1 · Beom‑Jin Lee2 · Cheong‑Weon Cho1

Received: 30 January 2017 / Accepted: 6 March 2017 / Published online: 3 April 2017
© The Korean Society of Pharmaceutical Sciences and Technology 2017

Abstract Drug release from a polymeric nanocarrier is Introduction


affected by several factors including the sort of composi-
tion (drug, polymer, and excipient), the ratio of composi- With the improvement of drug design methods, many
tion, physical or chemical interaction between components, potentially active substances have been manufactured and
and manufacturing methods. Depending on the mechanism synthesized. However, most recently developed drugs
of drug release from the vehicles, it can be divided into four belong to biopharmaceutical classification system (BCS)
categories (diffusion, solvent, chemical interaction, and groups 2 or 4, which have low aqueous solubility. Low
stimulated release). Recently, lipids have attracted great water solubility of drugs limits their absorption in the
interest as carriers for water-insoluble drug delivery. Lipid- body and reduces their oral bioavailability (Amidon et al.
based drug-delivery systems have received a lot of interest 1995). To make these drugs safe and effective for treat-
because of their ability to improve solubility and bioavail- ment, a number of factors including bioavailability, for-
ability of drugs that are poorly soluble in water. The lipid mulation characteristics, and body disposition must be
carrier, formulation strategy, and rational drug-delivery considered. Thus, attempts using micronization, formation
system should be selected appropriately for a lipid-based of complexes with cyclodextrin, solid dispersions, permea-
drug-delivery system to be successful. In this review, the tion enhancers, and surfactants have been made to solve
general release characteristics and mechanisms of drug the issues of dissolution and permeation of drugs (Aungst
from nanocarriers will be discussed. 1993). The goal of drug release from a carrier is to main-
tain and control the drug concentration in the blood and
Keywords Drug release · Polymeric nanocarrier · Lipid target tissue. The first paper describing a sustained drug-
nanoparticles · Release mechanism release system consisting of a polymeric device was pub-
lished in 1964 (Folkman and Long 1964), and a number of
delayed drug-delivery systems controlled by polymers have
begun to be investigated.
Several models of release kinetics can be used to illus-
trate drug release from drug-delivery systems, such as the
drug-release kinetic mechanisms controlled by the barrier
surrounding the matrix and regulated by drug diffusion
* Beom‑Jin Lee through a carrier matrix. In addition, degradation or swell-
bjl@ajou.ac.kr
ing of the carrier matrix and loss of drug–polymer linkage
* Cheong‑Weon Cho can also control the rate of drug release from the carrier
chocw@cnu.ac.kr
(Bajpai et al. 2008; Freiberg and; Zhu 2004). Recently,
1
College of Pharmacy, Chungnam National University, 99 various nanocarriers have been developed to improve the
Daehak‑ro, Yuseong‑gu, Daejeon 34134, Republic of Korea effectiveness of drug delivery (Lee and Yeo 2015) (Fig. 1).
2
College of Pharmacy, Ajou University, Suwon 16499, Although the spatial control of drug delivery based on
Republic of Korea nanocarriers has been widely studied (Acharya and Sahoo

13
Vol.:(0123456789)
288 G.-H. Son et al.

Fig. 1  Various types of phar- (a) (c)


(b)
maceutical nanocarriers for Hydrophobic
drug delivery. a nanocapsule; b drug
nanosphere; c liposome Drug

Hydrophilic
drug
Nanocapsule Nanosphere
Liposome

2011; Brannon-Peppas and Blanchette 2004; Lu and Low


2002; Maeda et al. 2013; Xu et al. 2013), the importance
of controlling drug release from carriers at a nanoscale is
sometimes neglected because large-scale drug-delivery sys-
tems are well established. However, because nanocarriers
have a larger surface area per volume and a short diffusion
distance, the control of drug release from nanoparticles
faces different challenges compared with classical drug
carriers.

Drug‑release mechanism from polymeric


nanocarriers

One of the primary goals of controlling drug release is


to keep the concentration of drug in the blood within the
therapeutic range (Siegel and Rathbone 2012). Therefore, Fig. 2  Various mechanisms of drug release from nanocarriers. a
it is ideal to develop drug carriers that have low dosing diffusion-controlled release; b solvent-controlled release; c polymer-
frequency and provide controlled drug release. To achieve degraded release; d pH-sensitive release
this, drug-delivery systems that have a zero-order drug-
release profile in which the drug is uniformly released have have membranes that can act as a barrier to diffusion. Thus,
been pursued (Bajpai et al. 2008; Siegel and Rathbone such systems generally show a high initial release, but over
2012). Drug release from a nanocarrier is affected by sev- time, the release rate decreases as the drug molecule diffu-
eral factors including the sort of composition (drug, poly- sion distance inside the carrier increases.
mer, and excipient), the ratio of composition, physical or
chemical interaction among components, and manufactur- Solvent‑controlled release
ing methods. Depending on the mechanism of drug release
out of the vehicles, drug release can be divided into four Transport of a solvent into drug-delivery systems may
categories (diffusion, solvent, chemical interaction, and affect drug-release behavior from the delivery carriers.
stimulated release), as shown in Fig. 2 (Langer and Peppas Solvent-controlled release includes osmotic- and swelling-
2006; Siegel and Rathbone 2012). controlled release (Langer and Peppas 2006). Osmotic-
controlled release occurs in a carrier that is packed with a
Diffusion‑controlled release semipermeable polymer membrane, and water flows from
the carrier with a low concentration of drug to the center of
Diffusion-controlled drug release occurs in capsule-like the carrier with high drug concentration. As a result of this
systems where the drug is dissolved or dispersed in a core mechanism, drug release with zero-order kinetics occurs
(Cauchetier et al. 2003). The diffusion of the drug is caused along a gradient of concentration that is constantly main-
by the difference in concentration gradient across the mem- tained across the membrane.
brane (Crank 1975). Here, the drug is dissolved in the A swelling-controlled system is mainly composed of
central part and then diffuses through the membrane. The polymer material having a three-dimensional cross-linked
matrix type nanospheres also have a diffusion-controlled network structure such as a hydrogel in which mesh size
release profile, where the drug molecules are evenly dis- controls drug-release behavior. (Lin and Metters 2006;
persed in the polymer matrix. Matrix type systems do not Peppas et al. 2000). Drug release from hydrogels can

13
Mechanisms of drug release from advanced drug formulations such as polymeric-based… 289

be analyzed by the semiempirical Peppas model ­ ( M t/ Drug‑release mechanisms of drug from lipid based
M∞ = ktn), where ­Mt and ­M∞ are the absolute accumula- nanocarriers
tions of drug released at time t and infinite time, respec-
tively, k is a constant, and n is the release index. This Recently, lipids have attracted a great deal of interest as
equation makes it possible to determine the release mech- carriers for water-insoluble drug delivery. The availabil-
anism (Hayashi et al. 2005; Korsmeyer et al. 1983; Pep- ity of useful lipid excipients capable of satisfying safety
pas et al. 2000; Ritger and Peppas 1987; Siepmann and aspects and having the ability to increase oral bioavail-
Peppas 2001). Swelling-controlled systems can achieve ability has contributed to the development of lipid-based
zero-order drug release, depending on the initial drug formulations. Lipid-based drug-delivery systems attract a
distribution of the system (Lee 1984) or polymer compo- lot of interest because of their ability to improve solubil-
sition (Kaity et al. 2013). ity and bioavailability of poorly water-soluble drugs (Pou-
ton 2006). Drug absorption from lipid-based formulations
depends on many factors including particle size, degree of
Degradation‑controlled release emulsification, rate of dispersion, and drug precipitation in
dispersion (Jannin et al. 2008; Pouton 2000, 2006). A lipid
Drug carriers composed of biodegradable polymers such carrier formulation strategy and a rational drug-delivery
as polyesters, polyamides, and polysaccharides release system should be selected appropriately for the lipid-based
the drug through enzymatic decomposition, which drug-delivery system to be successful (Dahan and Hoffman
degrades ester or amide bonds, or causes hydrolysis (Lee 2008).
et al. 2011; Prabaharan et al. 2009; Yoo and Park 2001).
A matrix composed of polymers such as polylactic-co-
glycolic acid (PLGA), polylactic acid (PLA), or polycap- Lipid based nanocarriers
rolactone (PCL) undergoes a degradation process, and
consequently the overall matrix is degraded simultane- Self‑micro emulsifying drug‑delivery systems
ously. By contrast, a matrix made from polymeric anhy- (SMEDDS)
drides or orthoesters typically erodes from the surface to
the center and then causes degradation of the polymer at SMEDDS are isotropic, transparent, and thermodynami-
a faster rate than water diffuses into the matrix (Burk- cally stable solutions containing oil, surfactant, and cosol-
ersroda et al. 2002; Middleton and Tipton 2000). How- vent/cosurfactant. SMEDDS are defined as systems capa-
ever, a small-sized matrix, such as those found in nano- ble of forming microemulsions of oil-in-water by simply
particles, has a very short diffusion length for water and a adding and gently agitating aqueous media such as gas-
limited crystallization zone. Polymer degradation contin- trointestinal (GI) fluids (Mahesh et al. 2001; Patel et al.
ues to accelerate with overall polymer degradation rather 2010). Microemulsion is characterized by small droplet
than only surface erosion (Lee and Chu 2008). Biode- size (about 200 nm) and wide interface. The drug is dis-
gradable polymer systems are preferred because they are solved in a droplet, and the large surface of the microemul-
degraded in the body. sion increases the absorption rate in the body. SMEDDS
not only promote solubilization of the drug but also have
an advantage in terms of releasing and absorbing the drug
pH‑controlled release (Craig et al. 1995; Farah and Denis 2001). Thus, if the
release rate of poorly water-soluble drugs acts as a key
The release of drugs from nanocarriers that respond to parameter for absorption, it is expected that SMEDDS will
stimuli is controlled by a stimulus such as temperature, improve the rate and extent of absorption, resulting in bet-
pH, ionic strength, ultrasound, electricity, or magnetic ter bioavailability (Gursoy and Benita 2004; Pouton 2000).
fields (Abouelmagd et al. 2014). Such carriers have been When SMEDDS are introduced by an oral route, emul-
studied for targeted specific drug delivery because it is sification occurs because of mixing with gastrointestinal
possible to localize stimulation. For example, nanocarri- fluids. Thus, the mixture of surfactant and oil containing
ers connected with pH-sensitive linkers using the weakly drugs is reconstituted to small-sized microemulsion drop-
acidic pH of many solid tumors have been developed lets. Figure 3 shows the various phases that will appear
as site-specific drug-delivery carriers (Min et al. 2010; when an aqueous medium is introduced into SMEDDS
Talelli et al. 2010). In heat-sensitive drug-delivery sys- (Rajput et al. 2012).
tems, drugs are released using the phase transition tem- The emulsified drug containing microemulsion drop-
perature of the heat-induced polymer (Chang et al. 2008; lets promotes bile secretion and is additionally emulsified
Li et al. 2011). by bile acid and bile salts. Figure 4 illustrates the process

13
290 G.-H. Son et al.

lipid bilayer. Figure 5 gives an overview of the various type


of liposomes that can be prepared by modifications, and the
models of drug-release kinetics from the liposome (Jain
and Jain 2016).
In general, drug release from liposomes depends on fac-
tors such as drug permeability and thermodynamic param-
eters such as drug distribution across bilayer surfaces. It is
known that in vitro sink conditions are intended to simulate
physiological states, and it is not easy to predict the com-
plexity of in vivo release processes (Jain and Jain 2016). To
establish better in vitro–in vivo correlations, release kinetic
studies were conducted in mathematical models with
appropriate approximation. Fugit and Anderson studied the
release patterns from liposomes in the nonsink environment
of topotecan, and they observed that drug release and distri-
bution between bilayer and aqueous phases followed first-
order release kinetics. Similar results were obtained after
a slight correction of the mathematical model when the
study was performed under sink conditions using dynamic
dialysis. Under nonsink conditions, mathematical modeling
was used to study the effect of drug dimerization and the
zeta potential on the drug distribution across bilayers (Fugit
and Anderson 2014). Dynamic dialysis is generally used to
study the kinetics of release from nanocarriers. Usually, the
drug is released from the nanocomposite and diffuses from
the dialysis membrane into the receiver portion of the sink
Fig. 3  The various phases that result from the addition of water condition. Control of permeability is an important param-
phase to SMEDDS eter in liposomes because it regulates active release, and
encapsulation serves to deliver the drug to the desired site
only. Drug-delivery systems based on liposomes may uti-
by which SMEDDS are absorbed after they enter the body. lize increased permeability of the phospholipid membrane
The lipid nanodroplets are metabolized by lipase of the through gel–sol transfer in response to external stimuli
pancreas and are divided into fatty acid and 2-monoglyc- (e.g., temperature causing drug release at the target site).
erides. Short fatty acids are absorbed by direct diffusion Although numerous studies have reported that liposomes
through the hepatic portal system, while long fatty acids can improve drug encapsulation and in vitro release, there
and monoglycerides are resynthesized into triglycerides exists no comprehensive theory on membrane structure and
and surrounded by phospholipids, cholesterol, and lipopro- drug release or membrane transport (Schaefer et al. 2012).
teins. As a result, chylomicrons eventually are created and Various dynamic models have been used to interpret drug
absorbed through the lymphatic pathway (Agrawal et al. release from liposomes (Costa and Sousa Lobo 2001; Fugit
2012). et al. 2015; Csuhai et al. 2015). There are various math-
ematical models for describing drug release from drug-
Liposomes delivery systems, including the zero-order kinetic model,
the first-order kinetic model, the Higuchi model, the Hix-
A liposome has a spherical bilayer structure similar to a cell son–Crowell model, the Korsmeyer–Peppas model, and the
membrane. The lipids predominantly used in the produc- regression model (Hayashi et al. 2005).
tion of liposomes are phospholipids having an amphipathic
hydrophilic head and a hydrophobic tail. These phospholip- Solid lipid nanoparticle (SLN)
ids form a spherical bilayer structure in the hydrated state,
with the hydrophobic portion facing inward and the hydro- Recently, SLN has received much attention as a means of
philic portion facing outward. The advantage of a liposome increasing site-specific drug delivery and bioavailability.
system is that the hydrophilic material can be buried in the Therefore, many studies have been conducted to explore the
hydrophilic space at the center of the liposome, and hydro- possibility of transport through the small intestinal lymph
phobic materials can be trapped in the fatty acid inside the pathway. SLN is a spherical particle typically 10–1000 nm

13
Mechanisms of drug release from advanced drug formulations such as polymeric-based… 291

Fig. 4  Potential mechanism for absorption enhancement of SMEDDS

in diameter, and the core of the solid lipid is stabilized by drug. In the cooling process of prepared oil-in-water type
surfactant and can solubilize lipid-soluble drugs and mate- nanoemulsions, the lower temperature of the aqueous phase
rials. One of the biggest challenges when studying lipid might reduce the solubility of drug. This means that the
nanoparticles is that burst release may occur in this system. drug is redistributed into the lipid. A schematic diagram of
A burst release phenomenon was observed in all encap- this process is shown in Fig. 6 (Muller et al. 2000). When
sulated drugs in tetracaine and etomidate SLNs prepared the lipid reaches a recrystallization temperature, the solid
by hot homogenization or cold homogenization methods, lipid core contains the drug. When the temperature of dis-
respectively (Schwarz 1995). By contrast, delayed release persion is lowered, the drug is redistributed to the lipid as
was obtained in the study of the incorporation of predni- the solubility of the drug in water is reduced. This is related
solone to SLN. These results confirm the suitability of the to the pressure exerted on the drug. Because the drug can
SLN system for long-term drug release. Importantly, it is no longer be present in the already crystallized lipid core,
possible to alter the drug-release profile by controlling the the drug is consequently concentrated on the surface of
lipid matrix, the concentration of the surfactant, and factors the SLN or the liquid outer layer. Thus, the extent of burst
involved in the production (e.g., temperature) of the lipid release can be controlled through the surfactant concentra-
nanoparticles, and the effect of particle size is negligible. tion or the temperature setting during preparation. Higher
In addition, manufacturing parameters such as surfactant temperature and surfactant concentration increase burst
concentration, temperature, and inherent characteristics of release, while production at room temperature does not
the lipid matrix are the greatest determinants of the drug- show any burst because it avoids the process of redistribu-
release profile from lipid nanoparticles. During nanopar- tion of the drug to the aqueous phase and subsequent redis-
ticle manufacturing processes involving high-temperature tribution to the lipid. SLN without burst release may be
homogenization techniques, the drug is distributed from a produced with a surfactant-free process. Based on Mehnert
liquid lipid phase to an aqueous phase. As the solubility of and Mader (2001), there are three drug encapsulation mod-
the drug increases in the aqueous phase, the amount of drug els in SLN; namely, a solid solution model, a core–shell
distributed to the aqueous phase will be greater. As temper- model, and a drug-enriched shell and core–shell model
ature and concentration of the surfactant increase, the drug (Fig. 6).
saturation solubility of the aqueous phase increases in a When lipid nanoparticles are prepared by cold homog-
system composed of water containing surfactant, lipid, and enization using no drug-solubilizing surfactant, the drug

13
292 G.-H. Son et al.

Fig. 5  Liposome modified in miscellaneous ways and various models of release kinetics from liposomes. a cationic or stimuli-sensitive lipo-
some; b release kinetics; c PEGylated or targeted liposomes; d conventional liposomes

is evenly dispersed on a molecular basis in the lipid is formed and surrounded by a lipid shell (Muller et al.
matrix, and this SLN matrix is a solid solution. The 2000).
model in which the drug is abundant in the outer shell
is because of drug redistribution during the cooling pro- Nanostructured lipid carrier (NLC)
cess. A drug-enriched core is obtained when the drug is
precipitated before the lipid is recrystallized. This core is Unlike SLN, where the drug is encapsulated in solid lipids,
obtained only when the drug is dissolved in the lipid at NLC refers to a system that encapsulates the drug in a mix-
or near saturation solubility. Cooling of the nanoemulsion ture of solid lipid and liquid lipid. In NLC, drug solubili-
allows the drug to crystallize before crystallization of the zation capacity increases because of liquid oil; thus, this
lipid, and the molten lipid will be supersaturated with the system exhibits controlled release characteristics with the
drug. Additional cooling will lead to recrystallization of advantage of high drug loading. In particular, incomplete
the lipid surrounding the drug core as a membrane. This and amorphous types of NLCs provide more flexibility
lipid membrane will contain only drug content corre- to achieve a desired sustained release (Fig. 7). NLC with
sponding to drug saturation solubility at lipid recrystal- more imperfections in the crystal structure as compared to
lization temperature. This means that a drug-rich core SLNs are prepared using a blend of solid lipid and spatially

13
Mechanisms of drug release from advanced drug formulations such as polymeric-based… 293

Fig. 6  Partitioning effects on drug during the production of SLN by a hot homogenization technique and three drug incorporation models [solid
solution model (left), drug-enriched shell models (middle) and lipid shell (right)]

shell and released by diffusion or erosion of the matrix. Fol-


lowing this initial rapid release, slow release appears from
the solid lipid core. The interesting aspect of NLC is that it
is possible to create a drug-release profile versus oil content
(Muller et al. 2002).

Drug‑release mechanisms

Peppas model

Korsmeyer et al. (1983) attempted to explain drug release as


a mathematical model (Korsmeyer–Peppas model) expressed
Fig. 7  Schematic illustration of SLN and NLC structure
in terms of a log cumulative drug-release percentage versus
log time. This model is stated as: M ­ t/M∞ = Ktη. ­Mt/M∞ is
different liquid lipids. These imperfections contribute to the fraction of drug released at time t, k is the release rate
improved drug loading and reduced drug expulsion during constant, and η is the release index. The η value predicts
storage (O’driscoll and Griffin 2008; Zhuang et al. 2010). the release mechanism of the drug. In other words, 0.45 ≤ η
Therefore, NLC which are lipid nanoparticles or colloidal corresponds to the Fickian diffusion model, 0.45 < η < 0.89
carriers have been explored as potential topical delivery to non-Fickian transport, η = 0.89 to Case II transport, and
vehicle. NLC have been reported to offer several advan- η > 0.89 to Super Case II transport.
tages over conventional topical products owing to their
ability to prolong the drug release, mitigate skin irritation, Higuchi model
and protect of drug from potential degradable opportuni-
ties. Additionally, the high specific surface area of the par- Higuchi (1963) has developed a number of mathematical
ticles ensures excellent contact with the affected site on the theoretical models to identify the manner in which water-
skin, facilitating the transfer of drug more efficiently (Fang soluble and lipid-soluble drugs are released from the various
et al. 2008). matrix systems. These models are represented by the follow-
NLC shows a drug-release pattern consisting of two ing equation:
stages: an initial burst release followed by a sustained release √
ft = Qt = D(2C − Cs)Ct
at a constant rate. The outer shell layer consisting of liquid
lipids is rich in the drug, which causes an initial burst release. where ­Qt is the amount of drug released per unit area at
Unlike SLN, the outer layer of NLC where liquid lipid is time t, C is the initial concentration of the drug, C
­ s is the
abundant can dissolve more lipophilic drugs. Thus, a consid- solubility of the drug, and D is the diffusion √
constant. The
erable amount of the drug can be easily loaded on the outer Higuchi model can be simplified to ft = KH t. ­KH is the

13
294 G.-H. Son et al.

Higuchi dissolution constant. Thus, Higuchi explained drug Two‑film theory mathematical model
release as a diffusion process based on Fick’s law (depend-
ing on the square root of time). In two-film theory mathematical models, interfacial
reactions and diffusion resistances are studied to evalu-
ate drug release and nanoparticle permeability from
First‑order release kinetics model
nanocrystals and liposomes. Small et al. (2012) used low-
frequency ultrasound (LFUS) to perturb the membrane.
The first-order kinetics model is expressed by the following
Liposomes were prepared from POPC (1-palmitoyl-2-ole-
equation: log C = log C ­ 0 – k­ t/2.303. ­C0 is the initial drug
oyl-sn-glycero-3-phosphocholine), dipalmitoylphosphati-
concentration, K is the first-order rate constant, and t is the
dylcholine (DPPC), and cholesterol. Calcein release from
time. The release data are shown as the cumulative log per-
large unilamellar vesicles (LUV) was monitored after
centage of remaining drug versus time and a straight line
LFUS (20 kHz) exposure following the two-film theo-
with a slope of K/2.303 (Dash et al. 2010, England et al.
retical mathematical model. Interestingly, the increased
2015). Three parameter models for liposomes have been
DPPC content increased permeability in response to
reported, which include reversible drug-delivery interac-
LFUS, while the permeability decreased when the molar
tions and first-order release kinetics. This model is simple
fraction of POPC and cholesterol increased.
and can be adapted to a wide range of nanocarriers using
different model parameters (Zeng et al. 2011).
Biomembrane model
Zero‑order release kinetics model
Biomembrane models simulate natural cell membranes
Slow drug release according to zero-order release kinetics because of similar lipid arrays (Sarpietro et al. 2013).
uses the following equation: Interactions between physiologically active compounds
and lipids have been studied using differential scanning
Dt = D0 + k0 t
calorimetry for a variety of purposes including drug
where ­Dt is the amount of drug dissolved at time t, ­D0 is the release from lipid vesicles to biomembrane models. The
initial drug amount in the solution, and ­k0 is the zero-order interaction between physiologically active substances and
release rate constant (Dash et al. 2010). In the zero-order biological membranes was observed using heat effect. In
release kinetics model, the drug-release data are expressed addition, various parameters (pH, swelling, and cross-
as the cumulative amount of released drug versus time. linking characteristics, etc.) have been found to influence
This release model can be useful for transdermal, oph- the release kinetics of bioactive molecules during DSC
thalmic, and poorly soluble drug delivery. The zero-order analysis (Sarpietro and Castelli 2011).
release pattern is ideal for slow and delayed drug delivery
such as antibiotics, antidepressants, blood pressure regula-
tors, analgesics, and anticancer drugs (Knepp et al. 1987; Toroidal model
Fattal et al. 1991).
For a liposome, the toroidal model has been well stud-
Weibull release model ied in peptide release (Torchilin and Lukyanov 2003).
For example, delta-lysin causes concentration gradient
The Weibull release rate model is an empirical model outflow of entrapped materials in phosphatidylcholine
widely used for both immediate and sustained drug-release (PC) vesicles. When fluorescence energy change was
patterns. Factors affecting overall drug release including used to study efflux effects and peptide-induced lipid flip-
effective surface area only depend on mass. This model is flops, peptide transitions across bilayers were observed
expressed as: with instantaneous agitation in the membrane. Sobko
et al. (2010) reported that the addition of the colicin E1
dw D channel-forming domain to liposomes results in a lipid
= CskMk� tp
dt h membrane penetration spread (flip-flop) with simultane-
where kM = S, and k′tp = X. ous release of fluorescent dye in the liposome. Colicin
Wei et al. (2014) developed a liposome containing bai- reflects the formation of a large pore that can induce the
calin to increase its oral bioavailability. This liposome release of colicin in liposomes and can be formed by the
showed delayed release according to the Weibull release head moiety of lipid molecules (Sobko et al. 2010).
rate model. In vivo studies have shown that oral bioavail-
ability increased threefold.

13
Mechanisms of drug release from advanced drug formulations such as polymeric-based… 295

Conclusions Crank J (1975) The mathematics of diffusion. Clarendon Press,


Oxford
Csuhai E, Kangarlou S, Xiang TX, Ponta A, Bummer P, Choi D,
The latest nanobased drug-delivery technology not only Anderson BD (2015) Determination of key parameters for a
provides increased solubility for poorly soluble drugs but mechanism-based model to predict Doxorubicin release from
also can control drug-release rate and pattern. Through actively loaded liposomes. J Pharm Sci 104:1087–1098
Dahan A, Hoffman A (2008) Rationalizing the selection of oral
studies of release mechanisms of nanocarrier into the body,
lipid based drug delivery systems by an in vitro dynamic lipol-
a true “controlled drug-delivery system” could be realized ysis model for improved oral bioavailability of poorly water
in the near future. soluble drugs. J Control Release 129:1–10
Dash S, Murthy PN, Nath L, Chowdhury P (2010) Kinetic mod-
Acknowledgements This work was supported by the Basic Sci- eling on drug release from controlled drug delivery systems.
ence Research Program (2016R1A2B4011294) through the National Acta Pol Pharm 67:217–223
Research Foundation of Korea (NRF) funded by the Ministry of Edu- Fang JY, Fang CL, Liu CH, Su YH (2008) Lipid nanoparticles as
cation, Science and Technology. vehicles for topical psoralen delivery: solid lipid nanoparti-
cles (SLN) versus nanostructured lipid carriers (NLC). Eur J
Compliance with ethical standards Pharm Biopharm 70:633–640
Farah N, Denis J (2001) Orally administrable composition capable
of providing enhanced bioavailability when ingested. US Pat-
Conflict of interest All authors declare that they have no conflict ent 6 ,312,704
of interest. Fattal E, Rojas J, Roblot-Treupel L, Andremont A, Couvreur P
(1991) Ampicillin-loaded liposomes and nanoparticles: com-
parison of drug loading, drug release and in vitro antimicrobial
activity. J Microencapsulation 8:29–36
Folkman J, Long DM (1964) The use of silicone rubber as a carrier
References for prolonged drug therapy. J Surg Res 4:139–142
Freiberg S, Zhu XX (2004) Polymer microspheres for controlled
Abouelmagd SA, Hyun H, Yeo Y (2014) Extracellularly activatable drug release. Int J Pharm 282:1–18
nanocarriers for drug delivery to tumors. Expert Opin Drug Fugit KD, Anderson BD (2014) Dynamic, nonsink method for the
Deliv 11:1601–1618 simultaneous determination of drug permeability and binding
Acharya S, Sahoo SK (2011) PLGA nanoparticles containing various coefficients in liposomes. Mol Pharm 11:1314–1325
anticancer agents and tumour delivery by EPR effect. Adv Drug Fugit KD, Xiang TX, Choi DH, Kangarlou S, Csuhai E, Bummer
Deliv Rev 63:170–183 PM, Anderson BD (2015) Mechanistic model and analysis of
Agrawal S, Giri TK, Tripathi DK, Alexander AA (2012) A review doxorubicin release from liposomal formulations. J Control
on novel therapeutics strategies for the enhancement of solubil- Release 217:82–91
ity for hydrophobic drugs through lipid and surfactant based self Gursoy RN, Benita S (2004) Self-emulsifying drug delivery sys-
micro emulsifying drug delivery system: a novel approach. Am J tems (SMEDDS) for improved oral delivery of lipophilic
Drug Discov Dev 2:143–183 drugs. Biomed Pharmacother 58:173–182
Amidon GL, Lennernas H, Shah VP, Crison JR (1995) A theoreti- Hayashi T, Kanbe H, Okada M, Suzuki M, Ikeda Y, Onuki Y,
cal basis for a biopharmaceutic drug classification: the correla- Kaneko T, Sonobe T (2005) Formulation study and drug
tion in vitro drug product dissolution and in vivo bioavailability. release mechanism of a new theophylline sustained-release
Pharm Res 12:413–420 preparation. Int J Pharm 304:91–101
Aungst BJ (1993) Novel formulation strategies for improving oral Higuchi T (1963) Mechanism of sustained-action medication.
bioavailability of drugs with poor membrane permeation or pre- Theoretical analysis of release of solid drug dispersed in solid
systemic metabolism. J Pharm Sci 82:979–987 matrices. J Pharm Sci 52:1145–1149
Bajpai AK, Shukla SK, Bhanu S, Kankane S (2008) Responsive poly- Jain A, Jain SK (2016) Stimuli-responsive smart liposomes in can-
mers in controlled drug delivery. Prog Polym Sci 33:1088–1118 cer targeting. Curr Drug Targets. doi:10.2174/1389450117666
Brannon-Peppas L, Blanchette JO (2004) Nanoparticle and targeted 160208144143
systems for cancer therapy. Adv Drug Deliv Rev 56:1649–1659 Jannin V, Musakhanian J, Marchaud D (2008) Approaches for the
Burkersroda FV, Schedl L, Göpferich A (2002) Why degradable development of solid and semi-solid lipid-based formulations.
polymers undergo surface erosion or bulk erosion. Biomaterials Adv Drug Deliv Rev 60:734–746
23:4221–4231 Kaity S, Isaac J, Ghosh A (2013) Interpenetrating polymer network
Cauchetier E, Deniau M, Fessi H, Astier A, Paul M (2003) of locust bean gum-poly (vinyl alcohol) for controlled release
Atovaquone-loaded nanocapsules: influence of the nature drug delivery. Carbohydr Polym 94:456–467
of the polymer on their in vitro characteristics. Int J Pharm Knepp VM, Hinz RS, Szoka FC, Guy RH (1987) Controlled drug
250:273–281 release from a novel liposomal delivery system. I. Investiga-
Chang C, Wei H, Quan CY, Li YY, Liu J, Wang ZC, Cheng SX, tion of transdermal potential. J Control Release 5:211–221
Zhang XZ, Zhuo RX (2008) Fabrication of thermosensitive Korsmeyer RW, Gurny R, Doelker E, Buri P, Peppas NA (1983)
PCL-PNIPAAm-PCL triblock copolymeric micelles for drug Mechanisms of potassium chloride release from compressed,
delivery. J Polym Sci Part A 46:3048–3057 hydrophilic, polymeric matrices: effect of entrapped air. J
Costa P, Sousa Lobo JM (2001) Modeling and comparison of dissolu- Pharm Sci 72:1189–1191
tion profiles. Eur J Pharm Sci 13:123–133 Langer R, Peppas N (2006) Chemical and physical structure of pol-
Craig DQM, Baker SA, Banning D, Booth SW (1995) An investiga- ymers as carriers for controlled release of bioactive agents: a
tion into the mechanisms of self-emulsification using particle review. J Macromol Sci 23:61–126
size analysis and low frequency dielectric spectroscopy. Int J
Pharm 114:103–110

13
296 G.-H. Son et al.

Lee PI (1984) Novel approach to zero-order drug delivery via immo- via pH-sensitive hydrazone bond for tumor-targeted drug deliv-
bilized nonuniform drug distribution in glass hydrogels. J Pharm ery. Biomaterials 30:5757–5766
Sci 73:1344–1347 Rajput DS, Alexander A, Jain V, Giri TK, Tripathi DK, Ajazuddin
Lee WC, Chu IM (2008) Preparation and degradation behavior of pol- (2012) Novel integrated approach for the strategic delivery of
yanhydrides nanoparticles. J Biomed Mater Res B 84:138–146 hydrophobic drugs by the use of self emulsifying drug delivery
Lee JH, Yeo Y (2015) Controlled drug release from pharmaceutical system. J Appl Sci 12(6):502–517
nanocarriers. Chem Eng Sci 125:75–84 Ritger PL, Peppas NA (1987) A simple equation for description of
Lee SH, Mok H, Lee Y, Park TG (2011) Self-assembled siRNA– solute release II. Fickian and anomalous release from swellable
PLGA conjugate micelles for gene silencing. J Control Release devices. J Control Release 5:37–42
152:152–158 Sarpietro MG, Castelli F (2011) Transfer kinetics from colloidal drug
Li W, Li J, Gao J, Li B, Xia Y, Meng Y, Yu Y, Chen H, Dai J, Wang carriers and liposomes to biomembrane models: DSC studies. J
H, Guo Y (2011) The fine-tuning of thermosensitive and degra- Pharm Bioall Sci 3:77–88
dable polymer micelles for enhancing intracellular uptake and Sarpietro MG, Accolla ML, Celia C, Grattoni A, Castelli F, Fresta M,
drug release in tumors. Biomaterials 32:3832–3844 Ferrari M, Paolino D (2013) Differential scanning calorimetry as
Lin CC, Metters AT (2006) Hydrogels in controlled release formu- a tool to investigate the transfer of anticancer drugs to biomem-
lations: network design and mathematical modeling. Adv Drug brane model. Curr Drug Targets 14:1053–1060
Deliv Rev 58:1379–1408 Schaefer JJ, Ma C, Harris JM (2012) Confocal Raman microscopy
Lu Y, Low PS (2002) Folate-mediated delivery of macromolecular probing of temperature-controlled release from individual, opti-
anticancer therapeutic agents. Adv Drug Deliv Rev 54:675–693 cally-trapped phospholipid vesicles. Anal Chem 84:9505–9512
Maeda H, Nakamura H, Fang J (2013) The EPR effect for macro- Schwarz C (1995) Lipidnanopartikel: herstellung, charakterisierung,
molecular drug delivery to solid tumors: improvement of tumor arzneistoffinkorporation and freisetzung, sterilization und lyoph-
uptake, lowering of systemic toxicity, and distinct tumor imaging ilisation. Ph.D. thesis, Free University of Berlin
in vivo. Adv Drug Deliv Rev 65:71–79 Siegel RA, Rathbone MJ (2012) Overview of controlled release
Mahesh D, Mandan J, Banode S (2001) Emulsion based drug delivery mechanisms. In: Siepmann J et al (eds) Fundamentals and appli-
system. Indian J Nov Drug Deliv 3:2–8 cations of controlled release drug delivery. Advances in delivery
Mehnert W, Mäder K (2001) Solid lipid nanoparticles: produc- science and technology. Springer, New York, pp 21–22
tion, characterization and applications. Adv Drug Deliv Rev Siepmann J, Peppas NA (2001) Modeling of drug release from deliv-
47:165–196 ery systems based on hydroxypropyl methylcellulose (HPMC).
Middleton JC, Tipton AJ (2000) Synthetic biodegradable polymers as Adv Drug Deliv Rev 48:139–157
orthopedic devices. Biomaterials 21:2335–2346 Small EF, Dan NR, Wrenn SP (2012) Low-frequency ultrasound-
Min KH, Kim JH, Bae SM, Shin H, Kim MS, Park S, Lee H, Park induced transport across non-raft-forming ternary lipid bilayers.
RW, Kim IS, Kim K, Kwon IC, Jeong SY, Lee DS (2010) Langmuir 28:14364–14372
Tumoral acidic pH- responsive MPEG-poly(β-amino ester)poly- Sobko AA, Kovalchuk SI, Kotova EA, Antonenko YN (2010) Induc-
meric micelles for cancer targeting therapy. J Control Release tion of lipid flip-flop colicin E1—a hallmark of proteolipidic pore
144:259–266 formation in liposome membranes. BioChemistry 75:728–733
Muller RH, Mader K, Gohla S (2000) Solid lipid nanoparticles (SLN) Talelli M, Iman M, Varkouhi AK, Rijcken CJF, Schiffelers RM,
for controlled drug delivery—a review of the state of the art. Eur Etrych T, Ulbrich, K, van Nostrum CF, Lammers T, Storm G,
J Pharm Biopharm 50:161–177 Hennink WE (2010) Core-cross linked polymeric micelles with
Muller RH, Radtke M, Wissing SA (2002) Soli lipid nanoparti- controlled release of covalently entrapped doxorubicin. Biomate-
cles (SLN) and nanostructured lipid carriers (NLC) in cos- rials 31:7797–7804
metic and dermatological preparations. Adv Drug Deliv Rev Torchilin VP, Lukyanov AN (2003) Peptide and protein drug delivery
54:S131–S155 to and into tumors: challenges and solutions. Drug Discov Today
O’driscoll CM, Griffin BT (2008) Biopharmaceutical challenges 8:259–266
associated with drugs with low aqueous solubility-the poten- Wei Y, Guo J, Zheng X, Wu J, Zhou Y, Yu Y, Ye Y, Zhang L, Zhao
tial impact of lipid-based formulations. Adv Drug Deliv Rev L (2014) Preparation, pharmacokinetics and biodistribution of
60:617–624 baicalin-loaded liposomes. Int J Nanomedicine 9:3623–3630
Patel PV, Desai RT, Kapupara PP (2010) Self emulsifying drug Xu S, Olenyuk BZ, Okamoto CT, Hamm-Alvarez SF (2013) Target-
delivery system: a conventional and alternative approach to ing receptor-mediated endocytotic pathways with nanoparticles:
improve oral bioavailability of lipophilic drugs. J Drug Dev Res rationale and advances. Adv Drug Deliv Rev 65:121–138
2:9344–9375 Yoo HS, Park TG (2001) Biodegradable polymeric micelles com-
Peppas NA, Bures P, Leobandung W, Ichikawa H (2000) Hydrogels in posed of doxorubicin conjugated PLGA–PEG blockcopolymer. J
pharmaceutical formulations. Eur J Pharm Biopharm 50:27–46 Control Release 70:63–70
Pouton CW (2000) Lipid formulations for oral administration of drugs Zeng L, An L, Wu X (2011) Modeling drug-carrier interac-
nanoemulsifying, self-emulsifying and self-microemulsifying tion in the drug release from nanocarriers. J Drug Deliv. doi:
drug delivery systems. Eur J Pharm Sci 11:S93–S98 10.1155/2011/370308
Pouton CW (2006) Formulation of poorly water-soluble drugs for Zhuang CY, Li N, Wang M, Zhang XN, Pan WS, Peng JJ, Pan YS,
oral administration: physicochemical and physiological issues Tang X (2010) Preparation and characterization of vinpocetine
and the lipid formulation classification system. Eur J Pharm Sci loaded nanostructured lipid carriers (NLC) for improved oral
29:278–287 bioavailability. Int J Pharm 394:179–185
Prabaharan M, Grailer JJ, Pilla S, Steeber DA, Gong S (2009) Amphi-
philic multi-arm-block copolymer conjugated with doxorubicin

13

You might also like