Pharmaceutical Dispersion Techniques For Dissolution and Bioavailability Enhancement of Poorly Water-Soluble Drugs
Pharmaceutical Dispersion Techniques For Dissolution and Bioavailability Enhancement of Poorly Water-Soluble Drugs
Pharmaceutical Dispersion Techniques For Dissolution and Bioavailability Enhancement of Poorly Water-Soluble Drugs
Review
Pharmaceutical Dispersion Techniques for
Dissolution and Bioavailability Enhancement of
Poorly Water-Soluble Drugs
Xingwang Zhang 1 ID
, Huijie Xing 2 , Yue Zhao 2 and Zhiguo Ma 1, *
1 Department of Pharmaceutics, College of Pharmacy, Jinan University, 601 West Huangpu Avenue,
Guangzhou 510632, China; zhangxw@jnu.edu.cn
2 Institute of Laboratory Animals, Jinan University, 601 West Huangpu Avenue, Guangzhou 510632, China;
huijie920326@163.com (H.X.); tsukimoon@126.com (Y.Z.)
* Correspondence: mzg1979@126.com
Received: 27 May 2018; Accepted: 19 June 2018; Published: 23 June 2018
Abstract: Over the past decades, a large number of drugs as well as drug candidates with poor
dissolution characteristics have been witnessed, which invokes great interest in enabling formulation
of these active ingredients. Poorly water-soluble drugs, especially biopharmaceutical classification
system (BCS) II ones, are preferably designed as oral dosage forms if the dissolution limit can be
broken through. Minimizing a drug’s size is an effective means to increase its dissolution and
hence the bioavailability, which can be achieved by specialized dispersion techniques. This article
reviews the most commonly used dispersion techniques for pharmaceutical processing that can
practically enhance the dissolution and bioavailability of poorly water-soluble drugs. Major interests
focus on solid dispersion, lipid-based dispersion (nanoencapsulation), and liquisolid dispersion
(drug solubilized in a non-volatile solvent and dispersed in suitable solid excipients for tableting or
capsulizing), covering the formulation development, preparative technique and potential applications
for oral drug delivery. Otherwise, some other techniques that can increase the dispersibility of a drug
such as co-precipitation, concomitant crystallization and inclusion complexation are also discussed.
Various dispersion techniques provide a productive platform for addressing the formulation challenge
of poorly water-soluble drugs. Solid dispersion and liquisolid dispersion are most likely to be
successful in developing oral dosage forms. Lipid-based dispersion represents a promising approach
to surmounting the bioavailability of low-permeable drugs, though the technique needs to traverse
the obstacle from liquid to solid transformation. Novel dispersion techniques are highly encouraged
to develop for formulation of poorly water-soluble drugs.
1. Introduction
Drugs with poor aqueous solubility are still an ongoing challenge in the successful formulation of
therapeutic products due to their low oral bioavailability. It is a hard nut to crack that has discouraged
pharmaceutical practitioners for many years. In the 1990s; the biopharmaceutical classification system
(BCS) was introduced to characterize various drugs according to their solubility and permeability [1].
It reports that over 70% of drugs and active entities are poorly water-soluble compounds (BCS II or BCS
IV) due to the considerable involvement of high throughput screening and combinatorial chemistry [2].
These active pharmaceutical ingredients (APIs) often suffer from formulation challenges because of
limited dissolution and/or low permeability. Accordingly; applicable formulation techniques are
highly aspired to improve the apparent solubility or dissolution of poorly soluble drugs and thus
enable them become bioavailable.
A variety of formulation strategies have been explored to overcome the poor aqueous solubility of
drugs, including micronization [3], nanocrystalization [4], salification [5], cyclodextrin inclusion [6,7],
cocrystallization [8], micelle solubilization [9], solid dispersion [10], liquisolid technique [11], and
encapsulation in nanoparticles [12]. Generally, there are two methods to prepare nanodrug, namely
top-down and bottom-up techniques. The former is a straightforward approach to reducing a drug’s
size by the mechanical force (grinding or crushing); the latter is a simple and self-dispersion process
where the drug is embedded or dissolved in carrier excipients/vehicles in molecular or amorphous
state by solubilization or self-assembly [13]. Of course, recrystallization from molecular solution by
antisolvent precipitation also represents a bottom-up technique for preparing drug nanocrystals [14].
However, the products produced by the top-down technique tend to result in broad size distribution
and insufficient physical stability due to potential Ostwald ripening, which limits its potential
application of this technique. The bottom-up technique, dispersion starting from molecules, almost can
maximize the dispersion of a drug and lead to more stable dispersion systems (amorphous, molecular
or colloidal). Therefore, the bottom-up dispersion technique represents the most promising approach
for pharmaceutical processing.
Pharmaceutical engineering involves all sorts of dispersion systems, including suspension system,
colloidal system and solution system, in which a drug can be dispersed by itself or in a solid matter,
a semisolid matter, a solvent or nanoparticles. Among these, solid dispersion (SDs), lipid-based
dispersion and liquisolid dispersion are well-developed and more commonly used pharmaceutical
dispersion techniques. These dispersion systems have been widely applied to formulation of poorly
water-soluble drugs to address the issues related to solubility and permeability. Solid dispersion
technology is a method of dispersing a drug in an inert carrier excipient (normally a water-soluble
polymer) in the solid form. This technique allows complete removal of drug crystallinity and molecular
dispersion of a poorly soluble drug in a hydrophilic polymeric carrier [15]. In solid dispersions, the
specific surface area of drug is dramatically amplified and the drug is always in high-energy state,
which greatly facilitates the dissolution. The performance of SDs as bioavailability enhancer has been
widely evaluated for different drugs with encouraging results [16]. Lipid-based formulations are a
series of preparations consisting of oils or lipids as basic excipients [17], in which the drug is highly
dispersed or completely solubilized in the lipophilic core. In addition to providing a supersaturated
drug concentration in the gastrointestinal tract, lipid formulations have the advantages of motivating
intestinal lymphatic drug transport and optimizing enterocyte-based drug transport and disposition
whereby to reduce the first-pass effect and increase the lipophilic drug’s absorption [18]. The potential
utility of lipid formulations as a means of bioavailability enhancer for poorly water-soluble drugs has
been critically reviewed [19,20].
Liquisolid system as a viable alternative to the conventionally used dispersion techniques for
dissolution and bioavailability improvement has received considerable attention in recent years [21].
Liquisolid formulations involve liquid medication in solid powdered form that possess a drug
delivery mechanism similar to soft capsules [22]. In the liquisolid systems, a drug is completely
dissolved in a non-volatile solvent and molecularly dispersed in suitable carrier and coating
materials. Liquisolid system breaks away from the constraint of Van der Waals’ attraction and
hydrophobic interaction between drug particulates, thus presenting the highest dispersibility and
physical stability among various dispersion systems. Better bioavailability for an orally administered
poorly water-soluble drug can be achieved through a liquisolid formulation since the drug is already in
solution. Of course, other dispersion techniques are also in active development, such as co-precipitation,
concomitant crystallization and inclusion complexation. These techniques provide flexible options to
tackle the low bioavailability of “problem” drugs caused by poor aqueous solubility.
In this article, a comprehensive survey on the use of solid dispersion, lipid-based solubilization
and liquisolid technique for dissolution and bioavailability enhancement of poor water-soluble
drugs are carried out. Specific aspects focus on the formulation development, excipient application,
preparative techniques and oral delivery efficiency of these dispersion-based formulations. In addition,
Pharmaceutics 2018, 10, x FOR PEER REVIEW 3 of 32
Pharmaceutics out.10,Specific
are carried2018, 74 aspects focus on the formulation development, excipient application, 3 of 33
Drug dissolution
Drug dissolutionrefers referstotoa aprocess
processthat that the
the interaction
interaction between
between drugdrug molecules
molecules is displaced
is displaced by
by the
the between
one one between the drugthe and
drugdissolution
and dissolution
medium medium [24].
[24]. If the If the intermolecular
intermolecular force betweenforcedrug
between drug
molecules
molecules is prematurely minimized, the dissolution will be significantly
is prematurely minimized, the dissolution will be significantly speeded up. Weak interactions between speeded up. Weak
interactions
drug and carrierbetween druginand
formed carrier formed
dispersions not onlyin dispersions
well maintain not only well maintain
the dispersion statethe
of dispersion
a drug in
state of a drug in a carrier, but also produce a higher internal energy
a carrier, but also produce a higher internal energy between drug and carrier than between drug between drug and carrier than
between drug molecules. This high-energy state greatly contributes
molecules. This high-energy state greatly contributes to the drug dissolution. Drug dissolution to the drug dissolution. Drug
dissolution
from from formulations
formulations is particularly is important
particularly forimportant
those drugs forwith
those drugsabsorption
a short with a short absorption
window, since
window, since they might have passed their absorptive sites
they might have passed their absorptive sites by the time they have dissolved. Dissolution by the time they have dissolved.
is the
Dissolution of
prerequisite is drug
the prerequisite
absorption by ofthe
drug absorption by
gastrointestinal the gastrointestinal
epithelia. Low aqueous epithelia. Low aqueous
solubility always results
solubility always results in a slow drug dissolution rate in the coarse
in a slow drug dissolution rate in the coarse dispersion systems. Formulating poorly soluble drugs dispersion systems. Formulating
poorly
into soluble
ultrafine drugs into
dispersions canultrafine
lower thedispersions
energy barrier canforlower the energy
dissolution barrier
in advance andfor dissolution
thus enhance the in
advance andrate.
dissolution thusForenhance
BCS IIthe dissolution
drugs, rate. For
it is a feasible wayBCStoIIpromote
drugs, ittheis aoral
feasible way toby
absorption promote
preparing the
oral absorption by preparing dissolution-unconfined dispersions.
dissolution-unconfined dispersions. However, for BCS IV drugs, it is not enough to improve the However, for BCS IV drugs, it is
not enough to improve the absorption extent utilizing a dispersion
absorption extent utilizing a dispersion strategy that merely overcomes the dissolution limit [25]. strategy that merely overcomes
the
It dissolution
must limit [25].
simultaneously It mustthe
surmount simultaneously
dissolution and surmount
absorption thebarriers.
dissolution and absorption
Formulation barriers.
techniques that
Formulation techniques that have the function of dispersion plus absorption-promoting
have the function of dispersion plus absorption-promoting effect are intrinsically required to develop. effect are
intrinsically required to develop. In this respect, lipid-based formulations
In this respect, lipid-based formulations have demonstrated great potential in absorption betterment have demonstrated great
potential
due to highinbiocompatibility
absorption betterment due towith
and interaction high thebiocompatibility
cell membrane [26]. andOfinteraction
course, lipid with the cell
dispersions,
membrane [26]. Of course, lipid dispersions, often in the form of
often in the form of nanoparticles, do not have to experience a dissolution process for subsequent nanoparticles, do not have to
experience aAnyhow,
absorption. dissolution process for
formulating subsequent
a poorly solubleabsorption. Anyhow,
drug into eligible formulating
dispersions can acreate
poorly soluble
favorable
drug into eligible dispersions can
conditions for its dissolution and absorption. create favorable conditions for its dissolution and absorption.
Pharmaceutics 2018, 10, 74 4 of 33
Pharmaceutics 2018, 10, x FOR PEER REVIEW 4 of 32
Figure 2.
Figure 2. Physical
Physicalstate
stateofofdrug
drugininsolid
soliddispersions:
dispersions:(a)(a)
molecular
molecularstate, forming
state, solid
forming solution;
solid (b)
solution;
amorphous
(b) amorphousstate, forming
state, forming amorphous
amorphous dispersions; (c)(c)microcrystal
dispersions; microcrystalstate,
state,forming
formingsimple
simpledispersions;
dispersions;
(d) colloidal state, forming colloidal dispersions.
(d) colloidal state, forming colloidal dispersions.
absorption of API, since the body preferably takes up the nutrients rather than the non-nutritive
excipients. Compared to low-molecular-weight carriers, polymeric carriers possess larger molecular
weight that can afford higher dispersibility and stronger recrystallization inhibition for drugs. For this
end, polymeric carriers are currently widely used for the preparation of SDs, such as PEG, PVP and
hydroxypropylmethylcellulose (HPMC). Nevertheless, the high viscosity, plasticity and hygroscopicity
associated with macromolecules that make problems for production compromise their application in
SDs. Polymeric carriers, not including surfactants, have inadequate absorption-promoting effect for
poorly permeable drugs, which just provide necessary dispersibility. Carriers that possess a surfactant
property, beyond dispersion powder, have the advantage of increasing drug absorption through
interaction with the absorptive epithelia and inhibiting drug efflux transporters. To improve the
performance of polymeric carriers, copolymers and functionalized polymers (e.g., PEGylated polymers)
are developed for SDs. These novel carrier excipients are provided with excellent amphiphilicity,
formability, solubilization or absorption-promoting characteristics. Examples include fatty acid
macrogolglycerides (e.g., Gelucire 44/14 and Gelucire 50/13), poly(vinylpyrrolidone-co-vinyl acetate)
(PVP/VA), and poly(vinyl acetate-co-vinyl caprolactame-co-ethylene glycol) (Soluplus® ).
Table 1. Summary of the commonly used excipients for preparation of solid dispersions.
Gelucire 44/14 and Gelucire 50/13 are non-ionic water dispersible surfactants composed
of well-characterized PEG-esters, a small glyceride fraction and free PEG, which are able to
self-emulsify upon contact with aqueous media, forming a fine dispersion [64]. The surface
activity of such PEGylated carriers can improve the solubility and wettability of API in vitro and
in vivo. Enhanced bioavailability was reported to be related with strong inhibition of P-glycoprotein
efflux and metabolizing enzyme CYP3A4 [65]. Furthermore, this kind of carriers have good
thermoplasticity suitable for use in the melt process. PVP/VA copolymers possesses proper flexibility,
Pharmaceutics 2018, 10, 74 6 of 33
bioadhesion, water
Pharmaceutics 2018, 10, xremoistenability
FOR PEER REVIEWand hardness, and have found usage in SDs as carrier excipient [60].
6 of 32
In comparison with PVP, PVP/VA has a lower hygroscopicity but higher bioadhesion, conferring it
processing
easy and good
processing and good absorption-promoting
absorption-promoting capacity. These
capacity. advantages
These advantages make it more
make suitable
it more for
suitable
preparation
for preparation of SDs
of SDsfor for
oraloral
drug delivery.
drug Soluplus
delivery. ®
Soluplus , a polyethylene
® , a polyethylene glycol, polyvinyl
glycol, acetate
polyvinyl and
acetate
polyvinylcaprolactame-based
and polyvinylcaprolactame-based graftgraft
copolymer, has has
copolymer, been extensively
been extensivelyinvestigated
investigated forfor
preparation
preparation of
SDs [62,63]. Soluplus ® is ®
a transparent solid excipient and can form solid
of SDs [62,63]. Soluplus is a transparent solid excipient and can form solid solutions with many solutions with many drugs
[66]. Soluplus
drugs ® is an®innovative excipient that enables new levels of solubility and bioavailability for
[66]. Soluplus is an innovative excipient that enables new levels of solubility and bioavailability
poorly soluble APIs. Soluplus
for poorly soluble APIs. Soluplus ® shows superior
® shows performance
superior performance in forming solidsolid
in forming solutions in theinhot
solutions themelt
hot
extrusion process thanks to its high flowability and excellent extrudability.
melt extrusion process thanks to its high flowability and excellent extrudability. The resulting solid The resulting solid
solution makes
solution makes API available in
API available in aa dissolved
dissolved state,
state, resulting
resulting in in significantly
significantly enhanced
enhanced bioavailability
bioavailability
in vivo. The products above-mentioned represents the latest development in carrier excipients of
in vivo. The products above-mentioned represents the latest development in carrier excipients of SDs,
SDs,
which will
which will become
become the the dominant
dominant excipients
excipients for
for production
production of of SDs
SDs inin the
the future
future[67].
[67].
Figure
Figure 3.
3. Commonly
Commonly used
used preparative techniques for
preparative techniques for solid
solid dispersions.
dispersions.
production. The most commonly used SCF for a variety of pharmaceutical applications is supercritical
carbon dioxide (CO2 ). Apart from lower critical temperature (Tc = 31.1 ◦ C) and pressure (Pc = 73.8 bar),
CO2 is nontoxic, nonflammable and inexpensive for use. In general, two basic SCF techniques can be
utilized to prepare SDs [74], namely rapid expansion of supercritical solution (RESS) and gas antisolvent
precipitation (GAS). In RESS technique, a solution containing drug and carrier in the supercritical state
is expanded rapidly through a nozzle. Due to rapid changes in density and solvent power, the solution
becomes highly supersaturated and consequently SDs are immediately formed [75]. In GAS technique,
drug and carrier are first dissolved in an organic solvent in a vessel. The solution is then pressurized
with a supercritical fluid, resulting in precipitation of the solid as a fine powder upon solvent extraction
by SCF [76]. Yin et al. prepared itraconazole SDs with HPMC, Pluronic F-127 and L-ascorbic acid
using GAS in an attempt to enhance its dissolution and bioavailability [77]. Powder X-ray diffraction
and Fourier transform infrared spectra indicated that itraconazole existed as an amorphous state in
SDs. SCF technology provides a novel alternative approach to preparing SDs with high surface area,
excellent flowability property and low solvent residue. This technology is equally applicable for scale-up
production of SDs and can avoid most of the drawbacks associated with the routine methods.
operated semi-continuously in a custom built device with the product throughput up to 1000 kg/h.
Therefore, the technique is incredibly applicable to the commercial processing of SDs.
4.2.2. Nanostructured
4.2.2. Lipid Carriers
Nanostructured Lipid Carriers
SLNs exclusively
SLNs exclusively involve
involve solid
solid lipids
lipids that
that have
have aa high
high crystallinity
crystallinity in
in the
the lipid
lipid core,
core, thus
thus existing
existing
potential drug expulsion upon storage [89]. The solid lipids typically exhibit low capacity
potential drug expulsion upon storage [89]. The solid lipids typically exhibit low capacity of dissolving of
dissolving
poorly poorly
soluble drugssoluble drugs
compared compared
with with In
liquid lipids. liquid lipids. Innanostructured
this context, this context, nanostructured lipid
lipid carriers (NLCs)
carriers (NLCs) are invented to overcome the limitations associated with SLNs owing to
are invented to overcome the limitations associated with SLNs owing to the highly ordered structure. the highly
ordered structure. NLCs are derived from SLNs by incorporating spatially incompatible liquid lipid
into the solid core. The participation of liquid lipid creates an imperfect crystal matrix, resulting in
higher drug loading and drug/lipid compatibility [90]. Depending on superb solubilizing and
Pharmaceutics 2018, 10, 74 11 of 33
NLCs are derived from SLNs by incorporating spatially incompatible liquid lipid into the solid core.
The participation of liquid lipid creates an imperfect crystal matrix, resulting in higher drug loading
and drug/lipid compatibility [90]. Depending on superb solubilizing and dispersing capacities, NLCs
have turned into a promising nanocarrier and have been widely investigated for oral drug delivery [91].
4.2.3. Nanoemulsions
Pharmaceutical nanoemulsions are generally O/W emulsions in the nanometer scale made up
of oil phase, water phase, emulsifier, and a selected co-emulsifier. By virtue of facile preparation and
smaller particle size, nanoemulsions have been getting considerable attention in recent years as smart
drug delivery system [92]. The oil used in the nanoemulsions formulation is a liquid lipid, which
provides a great practicality for high load of poorly soluble drugs. Nanoemulsions can spontaneously
form in the presence of massive surfactants (~20% of the oil phase, w/w) [93]. The excellence in
particle size (<100 nm) renders nanoemulsions a high dispersibility for drug and a great surface
area for absorption. Nanoemulsions can either be water-containing formulation or water-free
formulation. An example as water-free formulation is the self-microemulsifying drug delivery system
(SEDDS), generally being developed into the dosage form of soft capsules [94]. Drug dispersing
(molecularly dissolving) in the oil phase of nanoemulsions is equivalent to dispersing in the solid
carrier. They just appear in different state and dispersibility, but have no substantial distinction.
Therefore, nanoemulsions containing liquid lipid (oil) are also a kind of efficient dispersing vehicle.
motivating the pharmaceutical practitioners to develop various lipid-based formulations for coping
with challenges from compounds with inadequate solubility and permeability [107]. Apart from the
true solution and simple dosage forms, almost all colloidal dispersion systems are largely dependent
on the use of lipid excipients. For instance, in the formulation of emulsions, the hydrophilic drugs are
solubilized in the inner oil phase of emulsion droplets. In the case of liposomes, the hydrophilic drugs
are entrapped into the lipid bilayer of vesicles. In SLNs and NLCs, the hydrophilic drugs are dissolved
or dispersed in the lipid core of nanoparticles. For phytosomes, the natural compounds are physically
coupled or chemically conjugated with the phospholipids. Lipid excipients are generally inert, in vivo
biodegradable and biocompatible with the body, thus possessing high safety and accessibility for drug
delivery. Many kind of lipid excipients have been approved by the regulatory agency (Food and Drug
Administration, FDA) for use in pharmaceutical products [108]. Meanwhile, severe adverse reactions
have not been reported on lipid excipients or formulations as yet. Table 2 lists the commonly used
lipid excipients at large that are involved in the lipid-based formulations. With the advancement of
excipients, it will usher in the rapid development of lipid-based formulations to revitalize poorly
soluble and/or permeable drugs.
Table 2. Cont.
broccoli using 1-octanol as solvent in an attempt to further the intestinal permeability of NLCs [175].
The results showed that the intestinal permeability and bioavailability of tripterine were largely
improved through such functional NLCs. Nanoemulsions, another representative lipid nanocarrier,
have also demonstrated a great potential in promoting oral absorption of poorly water-soluble drugs.
Yin et al. developed biocompatible nanoemulsions using hemp oil and less surfactants for the oral
delivery of baicalein [93], and Chavez-Zamudio et al. prepared lysophosphatidylcholine-stabilized
nanoemulsions for the oral delivery of curcumin [176]. The constructed nanoemulsion systems
unexceptionally enhanced the oral bioavailability of payloads in comparison with their coarse
dispersions. A nanoemulsion system loading dabigatran etexilate phospholipid complex was also
proposed for use to improve the lipophilicity and oral bioavailability of drug [177]. In terms of
liposomes, there were also some reports related to their use in the oral delivery of poorly water-soluble
drugs [100]. As an example, Rushmi et al. formulated black seed oil (Nigella sativa) into liposomes
using the ethanol injection method aiming to enhance the oral bioavailability and improve the
therapeutic activity of such analgesic in small animal studies of analgesia [178]. The in vivo studies
showed that the liposomal formulation demonstrated a significant analgesic activity in mice. In recent
years, phytosomes (phospholipid complexes) are also being widely used for oral drug delivery [103].
For example, Freag et al. developed self-assembled phytosomal nanocarriers for improving the
solubility and oral bioavailability of celastrol [151], and Telange et al. developed apigenin-loaded
phytosomes to improve the drug’s aqueous solubility, dissolution, in vivo bioavailability, and
antioxidant activity [106]. It demonstrates that phytosome technique is a promising and viable
formulation strategy for enhancing the delivery efficiency of poorly water-soluble drugs.
Although lipid carriers have been proven to be potential as oral delivery vehicles, it should
be noted that the lipolysis of lipid carriers substantially takes place in the gastrointestinal transport
process. As a matter of fact, the in vivo degradation of lipid nanoparticles is not contradictory with their
absorption-promoting effect, since lipids can facilitate drug absorption by co-transport and cytosis in the
form of intact nanoparticles or reconstituted micelles [20]. The in vivo lipolysis-reconstitution mechanism
of lipid nanocarriers have been confirmed by Wu’s laboratory utilizing an environment-responsive
probe [179,180]. The fact that water-quenched fluorescent dye encapsulated in lipid nanoparticles can
be rekindled by the reconstitution of lipolytic products after lipolysis of nanoparticles provides pivotal
evidence for the gastrointestinal disposition of lipid nanocarriers.
nanosuspensions not only requires consideration for the formulation factors, but also the lyophilization
process. The process conditions have a crucial effect on the stability of nanoparticles during and after freeze
drying. During lyophilization, the nanoparticles will be subjected to various stresses, such as particles
agglomeration and desiccation, which may be detrimental to the stability of nanoparticles. Therefore, a
proper cryoprotectant and optimized lyophilization process must be adopted so as to minimize damage to
the nanostructures. For example, Howard et al. tested nine kinds of cryoprotectants and different freeze
drying conditions to optimize lyophilization process of solid lipid nanoparticles loading dexamethasone
palmitate for improving the long-term stability [183]. The resulting lyophilized SLNs exhibited slightly
larger but acceptable particle size and polydispersity index. Drug loading and particle shape were well
maintained by lyophilization. The lyophilized SLNs possessed a consistent particle size and less drug
content loss during a three month period. To improve the dissolution and intestinal permeability of
diosmin, Freag et al. utilized the lyophilization technique to prepare diosmin-loaded phytosomes [184].
The lyophilized phytosomal nanocarriers exhibited the lowest particle size to 316 nm, adequate ζ-potential
for stabilization of colloidal particles, and good in vitro stability. Phytosomes obtained by freeze drying
significantly improved the drug’s dissolution and permeation characteristics. Freeze drying is proven to
be an effective means to achieve a long-term stability of lipid nanocarriers.
4.5.3. Self-Emulsifying
Self-emulsifying drug delivery systems (SEDDS) are emulsifiable water-free formulations made
up of oil and emulsifier with or without hydrophilic co-solvent. SEDDS are utilized to solve
low bioavailability issues of poorly soluble and highly permeable drugs [189]. Self-emulsifying
formulations can rapidly disperse in the gastrointestinal tract in contact with the digestive fluids
under the agitation of gastrointestinal peristalsis to spontaneously form nanoemulsions, so-called
in situ self-emulsification. SEDDS are physically stable formulations and can be manufactured into the
semisolid soft capsules (e.g., Sandimmun® ) [190] or liquisolid tablets [191]. Water-free formulations
offer an opportunity to realize the commercial success of nanoemulsions.
Liquisolid compact tablets or capsules can be obtained after lipid dispersions are loaded into adsorptive
excipients and form dry-looking, freely flowing and compressible powders. For example, Nnamani et al.
developed low-dose liquisolid tablets of artemether-lumefantrine (AL) from NLCs and estimated their
Pharmaceutics
potential 2018,
for 10, delivery
oral x FOR PEERofREVIEW
AL in malariogenic Wistar mice [192]. The results highlighted 16 of 32
that
AL-loaded NLCs could be further processed into oral tablets to improve the patient’s compliance.
5. Liquisolid Dispersion Technique
5. Liquisolid Dispersion Technique
5.1. Overview of Liquisolid System
5.1. Overview of Liquisolid System
Molecular solution represents the highest dispersion of drug in a variety of formulations. In SDs,
Molecular
a poorly water solution
soluble represents
drug can form the highest dispersion
solid solution withof drug in a variety
a suitable carrier of formulations.
excipient. Solid
In SDs, a poorly water soluble drug can form solid solution with
pharmaceutical intermediates are readily processed into a final dosage form, such as tablets and a suitable carrier excipient.
Solid pharmaceutical
capsules. Accordingly,intermediates
there should be areareadily processed
possibility by which intoliquid
a final dosage form,
substances can be such as tablets
changed into
and solid
the capsules.
form.Accordingly, there should
Liquisolid dispersion be a possibility
technique by which
is the right means liquid substances solidification
to materialize can be changed of
into the solid form. Liquisolid dispersion technique is the right means to
liquefied drug. A liquisolid system refers to formulation implemented by conversion of lipophilic materialize solidification of
liquefied
drug in a drug. A liquisolid
nonvolatile solvent system refers to formulation
into dry-looking, implemented
freely flowing by conversion
and compressible of lipophilic
powders drug
by blending
aindrug
a nonvolatile
solutionsolvent into dry-looking,
with adsorptive freely
excipients flowing by
followed andprocessing
compressible intopowders
suitableby blending
dosage formsa drug
(as
solution with
illustrated adsorptive
in Figure excipients
5) [193]. followed by
The liquisolid processing
system greatlyinto suitablethe
maintains dosage forms (as
molecular illustrated
dispersion of
in Figure 5)
candidate [193].
drug andThe liquisolid
provides the system greatly maintains
most favorable dissolution thecondition,
molecularfreelydispersion of candidate
diffusing into the
drug and provides
dissolution medium.the most favorable
Therefore, dissolution
the liquisolid system condition,
possessesfreely diffusing
a number into the dissolution
of advantages: (1) drug
medium. Therefore,
dispersion in the solidthe liquisolid
formulation system possesses aliquid
as solubilized number of advantages:
form; (2) enabling (1)solidification
drug dispersion in the
of liquid
solid formulation
drug; as solubilized
(3) quicker drug dissolution liquid
fromform; (2) enabling
formulations duesolidification
to superior of liquid drug;
wettability and(3)miscibility;
quicker drug (4)
dissolution from formulations due to superior wettability and miscibility; (4)
low cost of production; and (5) liquisolid dispersions able to be developed either into immediately- low cost of production; and
(5) liquisolid
release or intodispersions able to be
sustained-release developed either
preparations. intothese
Besides immediately-release or into sustained-release
advantages, it requires the dose of drug
preparations.
for developing Besides these advantages,
a liquisolid system to beit relatively
requires the low dose of drugthat
in order for the
developing a liquisolid
drug solution can be system
fully
to be relatively
loaded within the lowsolid
in order thatLiquisolid
carriers. the drug solution
dispersion cantechnique
be fully loaded withinanthe
is becoming solid carriers.
innovative and
Liquisolid dispersion
promising formulation technique
strategyisthat
becoming an innovative
can improve and promising
the dissolution formulation strategy
and bioavailability of poorly that can
water
improve the dissolution and bioavailability of poorly water soluble drugs
soluble drugs [194]. Liquisolid dispersion technique is mainly implemented to enable the formulation[194]. Liquisolid dispersion
technique
of is mainlydrugs.
amphiphobic implemented to enable
These drugs tendthe to
formulation
precipitate of amphiphobic
from carriers, drugs.
evenThese
thoughdrugs tendare
they to
precipitate
temporarilyfrom carriers,ineven
entrapped though they are temporarily entrapped in nanoparticles.
nanoparticles.
Figure
Figure 5.5.Formulation
Formulationstrategy
strategy of poorly
of poorly waterwater soluble
soluble drugsthe
drugs using using the liquisolid
liquisolid dispersion dispersion
technique.
technique.
5.2. Formulation Components of Liquisolid System
5.2. Formulation Components of Liquisolid System
Liquisolid dispersion system is developed based on the principle of converting liquid medication into
freelyLiquisolid dispersion
flowing, readily systemand
compressible is apparently
developeddry based on the
powders principle
by physical of converting
adsorption liquid
using selected
medication into freely flowing, readily compressible and apparently dry powders by
excipients. In addition to common excipients involved in tablets or capsules, a liquisolid dispersion physical
adsorption usingconsists
system generally selected excipients. solvent,
of non-volatile In addition tomaterial
carrier common andexcipients involved
coating material. Theinformulation
tablets or
capsules, a liquisolid dispersion system generally consists of non-volatile solvent, carrier
composition of a typical liquisolid system (e.g., liquisolid compact tablets) is generalized in Table material
3.
and coating material. The formulation composition of a typical liquisolid system (e.g., liquisolid
compact tablets) is generalized in Table 3.
Pharmaceutics 2018, 10, 74 17 of 33
Non-volatile solvent
Non-volatile solventused usedin in liquisolid
liquisolid systems
systems should
should be inert,
be inert, lowlylowlyviscous, viscous, preferably
preferably water-
water-miscible and meanwhile have a strong solvent power as liquid
miscible and meanwhile have a strong solvent power as liquid vehicle. Various solvents with high vehicle. Various solvents with
high boiling
boiling point point are explored
are explored for thefor the formulation
formulation of liquisolid
of liquisolid systems, systems,
including including PEG, propylene
PEG, propylene glycol,
glycerin, and polysorbate. The non-volatile solvent simultaneously acts as a solventaand
glycol, glycerin, and polysorbate. The non-volatile solvent simultaneously acts as solvent and a
a binding
agent in the liquisolid formulation. It has been shown that the solvent had a significant effect on effect
binding agent in the liquisolid formulation. It has been shown that the solvent had a significant drug
on drug release from the liquisolid system [211]. For rapid-release purpose,
release from the liquisolid system [211]. For rapid-release purpose, a liquid vehicle in which the API a liquid vehicle in which
themost
is API soluble
is most is soluble
normallyis normally
selected.selected.
In the case In the case of sustained-release
of sustained-release preparation,preparation, the solvent
the solvent with a
with a highly viscosity is usually used to dissolve the drug, such as glycerin and Cremophor ® EL.
highly viscosity is usually used to dissolve the drug, such as glycerin and Cremophor EL. Of course, ®
also oftentimes
oftentimes involved
involved in theinliquisolid
the liquisolid systems.
systems.
In the
In theliquisolid
liquisolidsystem,
system, carrier
carrier materials
materials playplay the role
the key keyinrole in gaining
gaining the dry the formdry form of
of powders
powders from the solubilized liquid drug. Carrier materials mainly contribute
from the solubilized liquid drug. Carrier materials mainly contribute to liquid adsorption. It requires to liquid adsorption.
It requires
that the carrierthatshould
the carrier
be porous should and be porous
possess largeand possess
enough largesurface
specific enough area specific surface
(SSA) [212]. area
Carrier
(SSA) [212].
selection Carrier
depends selection
on its depends on
liquid adsorption its liquid
capacity, adsorption
flowability capacity,and
of powders flowability of powders
compressibility. The
most commonly used carriers in liquisolid formulations include MCC (e.g., Avicel® andMCC
and compressibility. The most commonly used carriers in liquisolid formulations include Ceolus(e.g.,
®),
Avicel ® and Ceolus® ), lactose, sorbitol, anhydrous dibasic calcium phosphate (Fujicalin® ), amorphous
lactose, sorbitol, anhydrous dibasic calcium phosphate (Fujicalin ), amorphous magnesium ®
magnesium aluminometasilicate ® ), etc. Among these, Fujicalin® and Neusilin® represent the
aluminometasilicate (Neusilin®),(Neusilin
etc. Among these, Fujicalin® and Neusilin® represent the newly-
newly-developed carrier materials,
developed carrier materials, which exhibit especiallywhich exhibit especially large
large SSA,SSA, upuptoto40 40 mm22/g/gand
and 300
300 m 2
m2/g,
/g,
respectively [210]. Neusilin ® not only can be used as a carrier material, but also function as a coating
respectively [210]. Neusilin not only can be used as a carrier material, but also function as a coating
®
material by
material by virtue
virtue of of excellent
excellent adsorption
adsorption and and flowability
flowability properties
properties [204].
[204].
Coating material
Coating material playsplays aa keykey role
role in
in covering
covering the the wet
wet carrier
carrier particles
particles to to form
form dry-looking
dry-looking and and
freely flowing powders. It should be a material possessing fine and
freely flowing powders. It should be a material possessing fine and highly adsorptive micropowders highly adsorptive micropowders
that can
that can adsorb
adsorb excess
excess liquid
liquid to to ensure
ensure goodgood flowability
flowability of of the
the admixture.
admixture. In In the
the liquisolid
liquisolid system,
system,
coating material
coating material and and carrier
carrier material
material mustmust be be used
used in in conjunction
conjunction in in order
order to to enhance
enhance the the flowing
flowing
and compressing properties of powders as illustrated in Figure
and compressing properties of powders as illustrated in Figure 6. Currently, the most commonly 6. Currently, the most commonly
used coating
coating material
material in in the ® and
used the liquisolid
liquisolid formulation
formulation is is colloidal
colloidal silicon
silicon dioxide,
dioxide, such such as as Aerosil
Aerosil® and
Cab-O-Sil®
Cab-O-Sil M5. Amorphous
® M5. Amorphous silica silica gel
gel (e.g., Syloid®
(e.g., Syloid Neusilin®®,,calcium
), Neusilin
®), calcium silicate (Florite®®)) and
silicate (Florite and ordered
ordered
mesoporous silicates that have suitable flowability and compressibility
mesoporous silicates that have suitable flowability and compressibility can also be used to prepare can also be used to prepare
liquisolid formulation
liquisolid formulation [11]. [11].
Figure
Figure 6.6.Formation process
Formation of dry-looking,
process freelyfreely
of dry-looking, flowing and compressible
flowing powderspowders
and compressible in the liquisolid
in the
system.
liquisolid system.
Apart from non-volatile solvent, carrier and coating materials, the liquisolid system usually use
Apart from non-volatile solvent, carrier and coating materials, the liquisolid system usually use
some other additives to develop solid dosage forms. Tablets or capsules normally experience the
some other additives to develop solid dosage forms. Tablets or capsules normally experience the
disintegration process before dissolution. Therefore, disintegrants such as sodium starch glycolate,
disintegration process before dissolution. Therefore, disintegrants such as sodium starch glycolate,
croscarmellose sodium, and low-substituted hydroxypropyl cellulose (L-HPC) are generally
croscarmellose sodium, and low-substituted hydroxypropyl cellulose (L-HPC) are generally included
included in liquisolid formulation to allow a fast disintegration. In addition, release modifier (e.g.,
in liquisolid formulation to allow a fast disintegration. In addition, release modifier (e.g., HPMC) and
HPMC) and crystal growth inhibitor (e.g., PVP) are frequently contained in the liquisolid compact
crystal growth inhibitor (e.g., PVP) are frequently contained in the liquisolid compact tablets [203,213].
tablets [203,213]. These additives improve the dissolution profile and physical stability of liquisolid
These additives improve the dissolution profile and physical stability of liquisolid systems.
systems.
with low drug crystallinity and higher dissolution rate (100% in 25 min). The oral bioavailability of
risperidone was significantly enhanced through liquisolid tablets in comparison to the conventional
tablets (1441.711 µg·h/mL vs. 321.011 µg·h/mL in AUC). The examples above indicate that liquisolid
technique was a potential tool in increasing dissolution and bioavailability of poorly water-soluble drugs.
7. Conclusions
To date, oral administration remains the most preferred route that patients take the
medicine, and solid dosage forms are the prevalent modality of pharmaceutical preparations.
Pharmaceutical processing always results in a dispersion of a drug in the solid, semisolid or liquid
excipient in particulate, colloidal, amorphous or molecular state. Super-advanced dispersion can
engender additional benefits for dissolution and bioavailability of poorly soluble drugs, which, of
course, greatly depends on effective dispersion techniques. It has been shown that solid dispersion,
lipid-based dispersion and liquisolid dispersion are becoming useful tools to solve the formulation
challenges of poorly soluble drugs.
Author Contributions: H.X. and Y.Z. collected the materials, X.Z. wrote the original manuscript and Z.M.
reviewed and edited the article.
Funding: This research was funded by the National Natural Science Foundation of China (81673604), the
Fundamental Research Funds for the Central Universities (21617473), the Natural Science Foundation of
Guangdong Province (2016A030310085) and the Scientific Research Cultivation and Innovation Foundation
of Jinan University (21616319; 21616317).
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Amidon, G.L.; Lennernas, H.; Shah, V.P.; Crison, J.R. A theoretical basis for a biopharmaceutic drug
classification: The correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm. Res.
1995, 12, 413–420. [CrossRef] [PubMed]
2. Goke, K.; Lorenz, T.; Repanas, A.; Schneider, F.; Steiner, D.; Baumann, K.; Bunjes, H.; Dietzel, A.; Finke, J.H.;
Glasmacher, B.; et al. Novel strategies for the formulation and processing of poorly water-soluble drugs.
Eur. J. Pharm. Biopharm. 2018, 126, 40–56. [CrossRef] [PubMed]
3. Leleux, J.; Williams, R.O., 3rd. Recent advancements in mechanical reduction methods: Particulate systems.
Drug Dev. Ind. Pharm. 2014, 40, 289–300. [CrossRef] [PubMed]
Pharmaceutics 2018, 10, 74 22 of 33
4. Scholz, P.; Keck, C.M. Nanocrystals: From raw material to the final formulated oral dosage form—A review.
Curr. Pharm. Des. 2015, 21, 4217–4228. [CrossRef] [PubMed]
5. He, Y.; Ho, C.; Yang, D.; Chen, J.; Orton, E. Measurement and accurate interpretation of the solubility of
pharmaceutical salts. J. Pharm. Sci. 2017, 106, 1190–1196. [CrossRef] [PubMed]
6. Zhang, X.; Zhang, T.; Lan, Y.; Wu, B.; Shi, Z. Nanosuspensions containing oridonin/hp-beta-cyclodextrin
inclusion complexes for oral bioavailability enhancement via improved dissolution and permeability.
AAPS PharmSciTech 2016, 17, 400–408. [CrossRef] [PubMed]
7. Ezawa, T.; Inoue, Y.; Murata, I.; Takao, K.; Sugita, Y.; Kanamoto, I. Characterization of the dissolution
behavior of piperine/cyclodextrins inclusion complexes. AAPS PharmSciTech 2018, 19, 923–933. [CrossRef]
[PubMed]
8. Gadade, D.D.; Pekamwar, S.S. Pharmaceutical cocrystals: Regulatory and strategic aspects, design and
development. Adv. Pharm. Bull. 2016, 6, 479–494. [CrossRef] [PubMed]
9. Cagel, M.; Tesan, F.C.; Bernabeu, E.; Salgueiro, M.J.; Zubillaga, M.B.; Moretton, M.A.; Chiappetta, D.A.
Polymeric mixed micelles as nanomedicines: Achievements and perspectives. Eur. J. Pharm. Biopharm. 2017,
113, 211–228. [CrossRef] [PubMed]
10. Vo, C.L.; Park, C.; Lee, B.J. Current trends and future perspectives of solid dispersions containing poorly
water-soluble drugs. Eur. J. Pharm. Biopharm. 2013, 85, 799–813. [CrossRef] [PubMed]
11. Vranikova, B.; Gajdziok, J. Liquisolid systems and aspects influencing their research and development.
Acta Pharm. 2013, 63, 447–465. [CrossRef] [PubMed]
12. Kalepu, S.; Nekkanti, V. Improved delivery of poorly soluble compounds using nanoparticle technology:
A review. Drug Deliv. Transl. Res. 2016, 6, 319–332. [CrossRef] [PubMed]
13. Kwok, P.C.; Chan, H.K. Nanotechnology versus other techniques in improving drug dissolution.
Curr. Pharm. Des. 2014, 20, 474–482. [CrossRef] [PubMed]
14. Mohammad, I.S.; He, W.; Yin, L. A Smart paclitaxel-disulfiram nanococrystals for efficient mdr reversal and
enhanced apoptosis. Pharm. Res. 2018, 35, 77. [CrossRef] [PubMed]
15. Meng, F.; Gala, U.; Chauhan, H. Classification of solid dispersions: Correlation to (i) stability and solubility
(ii) preparation and characterization techniques. Drug Dev. Ind. Pharm. 2015, 41, 1401–1415. [CrossRef]
[PubMed]
16. Fong, S.Y.; Bauer-Brandl, A.; Brandl, M. Oral bioavailability enhancement through supersaturation:
An update and meta-analysis. Expert Opin. Drug Deliv. 2017, 14, 403–426. [CrossRef] [PubMed]
17. Kalepu, S.; Manthina, M.; Padavala, V. Oral lipid-based drug delivery systems—An overview. Acta Pharm.
Sin. B 2013, 3, 361–372. [CrossRef]
18. Porter, C.J.H.; Trevaskis, N.L.; Charman, W.N. Lipids and lipid-based formulations: Optimizing the oral
delivery of lipophilic drugs. Nat. Rev. Drug Discov. 2007, 6, 231–248. [CrossRef] [PubMed]
19. Teixeira, M.C.; Carbone, C.; Souto, E.B. Beyond liposomes: Recent advances on lipid based nanostructures
for poorly soluble/poorly permeable drug delivery. Prog. Lipid Res. 2017, 68, 1–11. [CrossRef] [PubMed]
20. Xing, H.; Wang, H.; Wu, B.; Zhang, X. Lipid nanoparticles for the delivery of active natural medicines.
Curr. Pharm. Des. 2017, 23, 6705–6713. [CrossRef] [PubMed]
21. Gajdziok, J.; Vranikova, B. Enhancing of drug bioavailability using liquisolid system formulation.
Ceska Slov. Farm. 2015, 64, 55–66. [PubMed]
22. Azharshekoufeh, B.L.; Shokri, J.; Adibkia, K.; Javadzadeh, Y. Liquisolid technology: What it can do for
NSAIDs delivery? Colloids Surf. B Biointerfaces 2015, 136, 185–191. [CrossRef] [PubMed]
23. Zhang, X.; Sun, N.; Wu, B.; Lu, Y.; Guan, T.; Wu, W. Physical characterization of lansoprazole/PVP solid
dispersion prepared by fluid-bed coating technique. Powder Technol. 2008, 182, 480–485. [CrossRef]
24. Arthur, A.N.; Willis, R.W. The rate of dissolution of solid substances in their own solutions. J. Am. Chem. Soc.
1897, 19, 930–934.
25. Ghadi, R.; Dand, N. BCS class IV drugs: Highly notorious candidates for formulation development. J. Control.
Release 2017, 248, 71–95. [CrossRef] [PubMed]
26. Harde, H.; Das, M.; Jain, S. Solid lipid nanoparticles: An oral bioavailability enhancer vehicle. Expert Opin.
Drug Deliv. 2011, 8, 1407–1424. [CrossRef] [PubMed]
27. Xu, Y.; Liu, X.; Lian, R.; Zheng, S.; Yin, Z.; Lu, Y.; Wu, W. Enhanced dissolution and oral bioavailability
of aripiprazole nanosuspensions prepared by nanoprecipitation/homogenization based on acid-base
neutralization. Int. J. Pharm. 2012, 438, 287–295. [CrossRef] [PubMed]
Pharmaceutics 2018, 10, 74 23 of 33
28. Vasconcelos, T.; Sarmento, B.; Costa, P. Solid dispersions as strategy to improve oral bioavailability of poor
water soluble drugs. Drug Discov. Today 2007, 12, 1068–1075. [CrossRef] [PubMed]
29. Marano, S.; Barker, S.A.; Raimi-Abraham, B.T.; Missaghi, S.; Rajabi-Siahboomi, A.; Craig, D.Q.M.
Development of micro-fibrous solid dispersions of poorly water-soluble drugs in sucrose using
temperature-controlled centrifugal spinning. Eur. J. Pharm. Biopharm. 2016, 103, 84–94. [CrossRef] [PubMed]
30. Allen, L.V., Jr.; Yanchick, V.A.; Maness, D.D. Dissolution rates of corticosteroids utilizing sugar glass
dispersions. J. Pharm. Sci. 1977, 66, 494–497. [CrossRef] [PubMed]
31. Madgulkar, A.; Bandivadekar, M.; Shid, T.; Rao, S. Sugars as solid dispersion carrier to improve solubility
and dissolution of the BCS class II drug: Clotrimazole. Drug Dev. Ind. Pharm. 2016, 42, 28–38. [CrossRef]
[PubMed]
32. Martinez, L.M.; Videa, M.; Lopez Silva, T.; Castro, S.; Caballero, A.; Lara-Diaz, V.J.; Castorena-Torres, F.
Two-phase amorphous-amorphous solid drug dispersion with enhanced stability, solubility and
bioavailability resulting from ultrasonic dispersion of an immiscible system. Eur. J. Pharm. Biopharm.
2017, 119, 243–252. [CrossRef] [PubMed]
33. Das, A.; Nayak, A.K.; Mohanty, B.; Panda, S. Solubility and dissolution enhancement of etoricoxib by solid
dispersion technique using sugar carriers. ISRN Pharm. 2011, 2011, 819765. [CrossRef] [PubMed]
34. Duret, C.; Wauthoz, N.; Sebti, T.; Vanderbist, F.; Amighi, K. Solid dispersions of itraconazole for inhalation
with enhanced dissolution, solubility and dispersion properties. Int. J. Pharm. 2012, 428, 103–113. [CrossRef]
[PubMed]
35. Kauppinen, A.; Broekhuis, J.; Grasmeijer, N.; Tonnis, W.; Ketolainen, J.; Frijlink, H.W.; Hinrichs, W.L.J.
Efficient production of solid dispersions by spray drying solutions of high solid content using a 3-fluid
nozzle. Eur. J. Pharm. Biopharm. 2018, 123, 50–58. [CrossRef] [PubMed]
36. Thenmozhi, K.; Yoo, Y.J. Enhanced solubility of piperine using hydrophilic carrier-based potent solid
dispersion systems. Drug Dev. Ind. Pharm. 2017, 43, 1501–1509. [CrossRef] [PubMed]
37. Summers, M.P. Glass formation in barbiturates and solid dispersion systems of barbiturates with citric acid.
J. Pharm. Sci. 1978, 67, 1606–1610. [CrossRef] [PubMed]
38. Alam, M.A.; Al-Jenoobi, F.I.; Al-Mohizea, A.M.; Ali, R. Effervescence assisted fusion technique to enhance
the solubility of drugs. AAPS PharmSciTech 2015, 16, 1487–1494. [CrossRef] [PubMed]
39. Chawla, G.; Bansal, A.K. Improved dissolution of a poorly water soluble drug in solid dispersions with
polymeric and non-polymeric hydrophilic additives. Acta Pharm. 2008, 58, 257–274. [CrossRef] [PubMed]
40. Patil, M.P.; Gaikwad, N.J. Preparation and characterization of gliclazide-polyethylene glycol 4000 solid
dispersions. Acta Pharm. 2009, 59, 57–65. [PubMed]
41. Venkateskumar, K.; Parasuraman, S.; Gunasunderi, R.; Sureshkumar, K.; Nayak, M.M.; Shah, S.A.A.; Khoo, K.;
Kai, H.W. Acyclovir-polyethylene glycol 6000 binary dispersions: Mechanistic insights. AAPS PharmSciTech
2017, 18, 2085–2094. [CrossRef] [PubMed]
42. Silva, T.D.; Arantes, V.T.; Resende, J.A.; Speziali, N.L.; de Oliveira, R.B.; Vianna-Soares, C.D. Preparation and
characterization of solid dispersion of simvastatin. Drug Dev. Ind. Pharm. 2010, 36, 1348–1355. [CrossRef]
[PubMed]
43. Knopp, M.M.; Nguyen, J.H.; Becker, C.; Francke, N.M.; Jorgensen, E.B.; Holm, P.; Holm, R.; Mu, H.; Rades, T.;
Langguth, P. Influence of polymer molecular weight on in vitro dissolution behavior and in vivo performance
of celecoxib:PVP amorphous solid dispersions. Eur. J. Pharm. Biopharm. 2016, 101, 145–151. [CrossRef]
[PubMed]
44. Pradhan, R.; Tran, T.H.; Kim, S.Y.; Woo, K.B.; Choi, Y.J.; Choi, H.G.; Yong, C.S.; Kim, J.O. Preparation
and characterization of fast dissolving flurbiprofen and esomeprazole solid dispersion using spray drying
technique. Int. J. Pharm. 2016, 502, 38–46. [CrossRef] [PubMed]
45. Riekes, M.K.; Kuminek, G.; Rauber, G.S.; de Campos, C.E.; Bortoluzzi, A.J.; Stulzer, H.K. HPMC as a potential
enhancer of nimodipine biopharmaceutical properties via ball-milled solid dispersions. Carbohydr. Polym.
2014, 99, 474–482. [CrossRef] [PubMed]
46. Park, J.; Cho, W.; Cha, K.H.; Ahn, J.; Han, K.; Hwang, S.J. Solubilization of the poorly water soluble drug,
telmisartan, using supercritical anti-solvent (SAS) process. Int. J. Pharm. 2013, 441, 50–55. [CrossRef]
[PubMed]
Pharmaceutics 2018, 10, 74 24 of 33
47. Rashid, R.; Kim, D.W.; Din, F.U.; Mustapha, O.; Yousaf, A.M.; Park, J.H.; Kim, J.O.; Yong, C.S.; Choi, H.G.
Effect of hydroxypropylcellulose and Tween 80 on physicochemical properties and bioavailability of
ezetimibe-loaded solid dispersion. Carbohydr. Polym. 2015, 130, 26–31. [CrossRef] [PubMed]
48. Sarode, A.; Wang, P.; Cote, C.; Worthen, D.R. Low-viscosity hydroxypropylcellulose (HPC) grades SL and
SSL: Versatile pharmaceutical polymers for dissolution enhancement, controlled release, and pharmaceutical
processing. AAPS PharmSciTech 2013, 14, 151–159. [CrossRef] [PubMed]
49. Hirasawa, N.; Ishise, S.; Miyata, H.; Danjo, K. Application of nilvadipine solid dispersion to tablet formulation
and manufacturing using crospovidone and methylcellulose as dispersion carriers. Chem. Pharm. Bull. (Tokyo)
2004, 52, 244–247. [CrossRef] [PubMed]
50. Song, C.K.; Yoon, I.S.; Kim, D.D. Poloxamer-based solid dispersions for oral delivery of docetaxel: Differential
effects of F68 and P85 on oral docetaxel bioavailability. Int. J. Pharm. 2016, 507, 102–108. [CrossRef] [PubMed]
51. Balata, G.F.; Zidan, A.S.; Abourehab, M.A.; Essa, E.A. Rapid disintegrating tablets of simvastatin dispersions
in polyoxyethylene-polypropylene block copolymer for maximized disintegration and dissolution. Drug Des.
Dev. Ther. 2016, 10, 3211–3223. [CrossRef] [PubMed]
52. Karolewicz, B.; Nartowski, K.; Pluta, J.; Gorniak, A. Physicochemical characterization and dissolution studies
of acyclovir solid dispersions with Pluronic F127 prepared by the kneading method. Acta Pharm. 2016,
66, 119–128. [CrossRef] [PubMed]
53. Gorajana, A.; Kit, W.W.; Dua, K. Characterization and solubility study of norfloxacin-polyethylene glycol,
polyvinylpyrrolidone and carbopol 974p solid dispersions. Recent Patent. Drug Deliv. Formul. 2015, 9, 167–182.
[CrossRef]
54. Meng, F.; Meckel, J.; Zhang, F. Investigation of itraconazole ternary amorphous solid dispersions based on
povidone and Carbopol. Eur. J. Pharm. Sci. 2017, 106, 413–421. [CrossRef] [PubMed]
55. Liu, C.; Zhu, S.J.; Zhou, Y.; Wei, Y.P.; Pei, Y.Y. Enhancement of dissolution of cyclosporine A using solid
dispersions with polyoxyethylene (40) stearate. Pharmazie 2006, 61, 681–684. [PubMed]
56. Parmar, K.R.; Shah, S.R.; Sheth, N.R. Studies in dissolution enhancement of ezetimibe by solid dispersions in
combination with a surface adsorbent. Dissolut. Technol. 2011, 18, 55–61. [CrossRef]
57. Daravath, B.; Tadikonda, R.R.; Vemula, S.K. Formulation and pharmacokinetics of gelucire solid dispersions
of flurbiprofen. Drug Dev. Ind. Pharm. 2015, 41, 1254–1262. [CrossRef] [PubMed]
58. Panda, T.; Das, D.; Panigrahi, L. Formulation development of solid dispersions of bosentan using Gelucire
50/13 and Poloxamer 188. J. Appl. Pharm. Sci. 2016, 027–033. [CrossRef]
59. Bali, D.E.; Osman, M.A.; El Maghraby, G.M. Enhancement of dissolution rate and intestinal stability of
clopidogrel hydrogen sulfate. Eur. J. Drug Metab. Pharmacokinet. 2016, 41, 807–818. [CrossRef] [PubMed]
60. Song, Y.; Wang, L.; Yang, P.; Wenslow, R.M., Jr.; Tan, B.; Zhang, H.; Deng, Z. Physicochemical characterization
of felodipine-kollidon VA64 amorphous solid dispersions prepared by hot-melt extrusion. J. Pharm. Sci.
2013, 102, 1915–1923. [CrossRef] [PubMed]
61. Knopp, M.M.; Nguyen, J.H.; Mu, H.; Langguth, P.; Rades, T.; Holm, R. Influence of copolymer composition
on in vitro and in vivo performance of celecoxib-PVP/VA amorphous solid dispersions. AAPS J. 2016,
18, 416–423. [CrossRef] [PubMed]
62. Ogawa, N.; Hiramatsu, T.; Suzuki, R.; Okamoto, R.; Shibagaki, K.; Fujita, K.; Takahashi, C.; Kawashima, Y.;
Yamamoto, H. Improvement in the water solubility of drugs with a solid dispersion system by spray drying
and hot-melt extrusion with using the amphiphilic polyvinyl caprolactam-polyvinyl acetate-polyethylene
glycol graft copolymer and d-mannitol. Eur. J. Pharm. Sci. 2018, 111, 205–214. [CrossRef] [PubMed]
63. Kulkarni, C.; Kelly, A.L.; Gough, T.; Jadhav, V.; Singh, K.K.; Paradkar, A. Application of hot melt extrusion
for improving bioavailability of artemisinin a thermolabile drug. Drug Dev. Ind. Pharm. 2018, 44, 206–214.
[CrossRef] [PubMed]
64. Swain, R.P.; Subudhi, B.B. Effect of semicrystalline polymers on self-emulsifying solid dispersions of
nateglinide: In vitro and in vivo evaluation. Drug Dev. Ind. Pharm. 2018, 44, 56–65. [CrossRef] [PubMed]
65. Wahlang, B.; Kabra, D.; Pawar, Y.B.; Tikoo, K.; Bansal, A.K. Contribution of formulation and excipients
towards enhanced permeation of curcumin. Arzneimittelforschung 2012, 62, 88–93. [CrossRef] [PubMed]
66. Grymonpre, W.; Bostijn, N.; Herck, S.V.; Verstraete, G.; Vanhoorne, V.; Nuhn, L.; Rombouts, P.; Beer, T.;
Remon, J.P.; Vervaet, C. Downstream processing from hot-melt extrusion towards tablets: A quality by
design approach. Int. J. Pharm. 2017, 531, 235–245. [CrossRef] [PubMed]
Pharmaceutics 2018, 10, 74 25 of 33
67. Vasconcelos, T.; Marques, S.; das Neves, J.; Sarmento, B. Amorphous solid dispersions: Rational selection of
a manufacturing process. Adv. Drug Deliv. Rev. 2016, 100, 85–101. [CrossRef] [PubMed]
68. Sekiguchi, K.; Obi, N. Studies on absorption of eutectic mixtures. I. A comparison of the behavior of eutectic
mixtures of sulphathiazole and that of ordinary sulphathiazole in man. Chem. Pharm. Bull. 1961, 9, 866–872.
[CrossRef]
69. Alhayali, A.; Tavellin, S.; Velaga, S. Dissolution and precipitation behavior of ternary solid dispersions of
ezetimibe in biorelevant media. Drug Dev. Ind. Pharm. 2017, 43, 79–88. [CrossRef] [PubMed]
70. Dos Santos, K.M.; Barbosa, R.M.; Vargas, F.G.A.; de Azevedo, E.P.; Lins, A.; Camara, C.A.; Aragao, C.F.S.;
Moura, T.; Raffin, F.N. Development of solid dispersions of beta-lapachone in PEG and PVP by solvent
evaporation method. Drug Dev. Ind. Pharm. 2017, 44, 750–756. [CrossRef] [PubMed]
71. Chen, Y.; Shi, Q.; Chen, Z.; Zheng, J.; Xu, H.; Li, J.; Liu, H. Preparation and characterization of emulsified
solid dispersions containing docetaxel. Arch. Pharm. Res. 2011, 34, 1909–1917. [CrossRef] [PubMed]
72. Nokhodchi, A.; Talari, R.; Valizadeh, H.; Jalali, M.B. An investigation on the solid dispersions of
chlordiazepoxide. Int. J. Biomed. Sci. 2007, 3, 211–216. [PubMed]
73. Weuts, I.; Kempen, D.; Verreck, G.; Decorte, A.; Heymans, K.; Peeters, J.; Brewster, M.; Van den Mooter, G.
Study of the physicochemical properties and stability of solid dispersions of loperamide and PEG6000
prepared by spray drying. Eur. J. Pharm. Biopharm. 2005, 59, 119–126. [CrossRef] [PubMed]
74. Naylor, A.; Lewis, A.L.; Ilium, L. Supercritical fluid-mediated methods to encapsulate drugs: Recent advances
and new opportunities. Ther. Deliv. 2011, 2, 1551–1565. [CrossRef] [PubMed]
75. Hezave, A.Z.; Aftab, S.; Esmaeilzadeh, F. Micronization of creatine monohydrate via Rapid Expansion of
Supercritical Solution (RESS). J. Supercrit. Fluids 2010, 55, 316–324. [CrossRef]
76. Li, S.; Liu, Y.; Liu, T.; Zhao, L.; Zhao, J.; Feng, N. Development and in-vivo assessment of the bioavailability
of oridonin solid dispersions by the gas anti-solvent technique. Int. J. Pharm. 2011, 411, 172–177. [CrossRef]
[PubMed]
77. Yin, X.; Daintree, L.S.; Ding, S.; Ledger, D.M.; Wang, B.; Zhao, W.; Qi, J.; Wu, W.; Han, J. Itraconazole
solid dispersion prepared by a supercritical fluid technique: Preparation, in vitro characterization, and
bioavailability in beagle dogs. Drug Des. Dev. Ther. 2015, 9, 2801–2810. [PubMed]
78. LaFountaine, J.S.; McGinity, J.W.; Williams, R.O., 3rd. Challenges and strategies in thermal processing of
amorphous solid dispersions: A review. AAPS PharmSciTech 2016, 17, 43–55. [CrossRef] [PubMed]
79. LaFountaine, J.S.; Jermain, S.V.; Prasad, L.K.; Brough, C.; Miller, D.A.; Lubda, D.; McGinity, J.W.;
Williams, R.O., 3rd. Enabling thermal processing of ritonavir-polyvinyl alcohol amorphous solid dispersions
by KinetiSol® Dispersing. Eur. J. Pharm. Biopharm. 2016, 101, 72–81. [CrossRef] [PubMed]
80. Rumondor, A.C.; Dhareshwar, S.S.; Kesisoglou, F. Amorphous solid dispersions or prodrugs: Complementary
strategies to increase drug absorption. J. Pharm. Sci. 2016, 105, 2498–2508. [CrossRef] [PubMed]
81. Deng, L.; Wang, Y.; Gong, T.; Sun, X.; Zhang, Z.R. Dissolution and bioavailability enhancement of
alpha-asarone by solid dispersions via oral administration. Drug Dev. Ind. Pharm. 2017, 43, 1817–1826.
[CrossRef] [PubMed]
82. Thiry, J.; Kok, M.G.; Collard, L.; Frere, A.; Krier, F.; Fillet, M.; Evrard, B. Bioavailability enhancement of
itraconazole-based solid dispersions produced by hot melt extrusion in the framework of the Three Rs rule.
Eur. J. Pharm. Sci. 2017, 99, 1–8. [CrossRef] [PubMed]
83. Otsuka, M.; Maeno, Y.; Fukami, T.; Inoue, M.; Tagami, T.; Ozeki, T. Solid dispersions of efonidipine
hydrochloride ethanolate with improved physicochemical and pharmacokinetic properties prepared with
microwave treatment. Eur. J. Pharm. Biopharm. 2016, 108, 25–31. [CrossRef] [PubMed]
84. Ye, Y.; Zhang, X.; Zhang, T.; Wang, H.; Wu, B. Design and evaluation of injectable niclosamide nanocrystals
prepared by wet media milling technique. Drug Dev. Ind. Pharm. 2015, 41, 1416–1424. [CrossRef] [PubMed]
85. Liu, Y.; Salituro, G.M.; Lee, K.J.; Bak, A.; Leung, D.H. Modulating drug release and enhancing the
oral bioavailability of torcetrapib with solid lipid dispersion formulations. AAPS PharmSciTech 2015,
16, 1091–1100. [CrossRef] [PubMed]
86. Hu, X.; Dong, X.; Lu, Y.; Qi, J.; Zhao, W.; Wu, W. Bioimaging of nanoparticles: The crucial role of
discriminating nanoparticles from free probes. Drug Discov. Today 2017, 22, 382–387. [CrossRef] [PubMed]
87. Muller, R.H.; Mader, K.; Gohla, S. Solid lipid nanoparticles (SLN) for controlled drug delivery—A review of
the state of the art. Eur. J. Pharm. Biopharm. 2000, 50, 161–177. [CrossRef]
Pharmaceutics 2018, 10, 74 26 of 33
88. Muchow, M.; Maincent, P.; Muller, R.H. Lipid nanoparticles with a solid matrix (SLN, NLC, LDC) for oral
drug delivery. Drug Dev. Ind. Pharm. 2008, 34, 1394–1405. [CrossRef] [PubMed]
89. Zhang, Y.; Li, Z.; Zhang, K.; Yang, G.; Wang, Z.; Zhao, J.; Hu, R.; Feng, N. Ethyl oleate-containing
nanostructured lipid carriers improve oral bioavailability of trans-ferulic acid ascompared with conventional
solid lipid nanoparticles. Int. J. Pharm. 2016, 511, 57–64. [CrossRef] [PubMed]
90. Andrade, L.M.; de Fatima Reis, C.; Maione-Silva, L.; Anjos, J.L.; Alonso, A.; Serpa, R.C.; Marreto, R.N.;
Lima, E.M.; Taveira, S.F. Impact of lipid dynamic behavior on physical stability, in vitro release and skin
permeation of genistein-loaded lipid nanoparticles. Eur. J. Pharm. Biopharm. 2014, 88, 40–47. [CrossRef]
[PubMed]
91. Poonia, N.; Kharb, R.; Lather, V.; Pandita, D. Nanostructured lipid carriers: Versatile oral delivery vehicle.
Future Sci. OA 2016, 2, Fso135. [CrossRef] [PubMed]
92. Aboalnaja, K.O.; Yaghmoor, S.; Kumosani, T.A.; McClements, D.J. Utilization of nanoemulsions to enhance
bioactivity of pharmaceuticals, supplements, and nutraceuticals: Nanoemulsion delivery systems and
nanoemulsion excipient systems. Expert Opin. Drug Deliv. 2016, 13, 1327–1336. [CrossRef] [PubMed]
93. Yin, J.; Xiang, C.; Wang, P.; Yin, Y.; Hou, Y. Biocompatible nanoemulsions based on hemp oil and less
surfactants for oral delivery of baicalein with enhanced bioavailability. Int. J. Nanomed. 2017, 12, 2923–2931.
[CrossRef] [PubMed]
94. Gursoy, R.N.; Benita, S. Self-emulsifying drug delivery systems (SEDDS) for improved oral delivery of
lipophilic drugs. Biomed. Pharmacother. 2004, 58, 173–182. [CrossRef] [PubMed]
95. Bulbake, U.; Doppalapudi, S.; Kommineni, N.; Khan, W. Liposomal formulations in clinical use: An updated
review. Pharmaceutics 2017, 9. [CrossRef] [PubMed]
96. Zhang, X.; Qi, J.; Lu, Y.; He, W.; Li, X.; Wu, W. Biotinylated liposomes as potential carriers for the oral delivery
of insulin. Nanomedicine 2014, 10, 167–176. [CrossRef] [PubMed]
97. Deng, W.; Xie, Q.; Wang, H.; Ma, Z.; Wu, B.; Zhang, X. Selenium nanoparticles as versatile carriers for
oral delivery of insulin: Insight into the synergic antidiabetic effect and mechanism. Nanomedicine 2017,
13, 1965–1974. [CrossRef] [PubMed]
98. Sauvage, F.; Franze, S.; Bruneau, A.; Alami, M.; Denis, S.; Nicolas, V.; Lesieur, S.; Legrand, F.X.; Barratt, G.;
Messaoudi, S.; et al. Formulation and in vitro efficacy of liposomes containing the Hsp90 inhibitor 6BrCaQ
in prostate cancer cells. Int. J. Pharm. 2016, 499, 101–109. [CrossRef] [PubMed]
99. Monteiro, L.O.F.; Fernandes, R.S.; Oda, C.M.R.; Lopes, S.C.; Townsend, D.M.; Cardoso, V.N.; Oliveira, M.C.;
Leite, E.A.; Rubello, D.; de Barros, A.L.B. Paclitaxel-loaded folate-coated long circulating and pH-sensitive
liposomes as a potential drug delivery system: A biodistribution study. Biomed. Pharmacother. 2017,
97, 489–495. [CrossRef] [PubMed]
100. Daeihamed, M.; Dadashzadeh, S.; Haeri, A.; Akhlaghi, M.F. Potential of liposomes for enhancement of oral
drug absorption. Curr. Drug Deliv. 2017, 14, 289–303. [CrossRef] [PubMed]
101. Jain, N.; Gupta, B.P.; Thakur, N.; Jain, R.; Banweer, J.; Jain, D.K.; Jain, S. Phytosome: A novel drug delivery
system for herbal medicine. Int. J. Pharm. Sci. Drug Res. 2010, 2, 224–228.
102. Matias, D.; Rijo, P.; Reis, C.P. Phytosomes as biocompatible carriers of natural drugs. Curr. Med. Chem. 2017,
24, 568–589. [CrossRef] [PubMed]
103. Gnananath, K.; Sri Nataraj, K.; Ganga Rao, B. Phospholipid complex technique for superior bioavailability of
phytoconstituents. Adv. Pharm. Bull. 2017, 7, 35–42. [CrossRef] [PubMed]
104. El-Gazayerly, O.N.; Makhlouf, A.I.; Soelm, A.M.; Mohmoud, M.A. Antioxidant and hepatoprotective
effects of silymarin phytosomes compared to milk thistle extract in CCl4 induced hepatotoxicity in rats.
J. Microencapsul. 2014, 31, 23–30. [CrossRef] [PubMed]
105. Mirzaei, H.; Shakeri, A.; Rashidi, B.; Jalili, A.; Banikazemi, Z.; Sahebkar, A. Phytosomal curcumin: A review
of pharmacokinetic, experimental and clinical studies. Biomed. Pharmacother. 2017, 85, 102–112. [CrossRef]
[PubMed]
106. Telange, D.R.; Patil, A.T.; Pethe, A.M.; Fegade, H.; Anand, S.; Dave, V.S. Formulation and characterization
of an apigenin-phospholipid phytosome (APLC) for improved solubility, in vivo bioavailability, and
antioxidant potential. Eur. J. Pharm. Sci. 2017, 108, 36–49. [CrossRef] [PubMed]
107. Mu, H.; Holm, R.; Mullertz, A. Lipid-based formulations for oral administration of poorly water-soluble
drugs. Int. J. Pharm. 2013, 453, 215–224. [CrossRef] [PubMed]
Pharmaceutics 2018, 10, 74 27 of 33
108. Chen, M.L. Lipid excipients and delivery systems for pharmaceutical development: A regulatory perspective.
Adv. Drug Deliv. Rev. 2008, 60, 768–777. [CrossRef] [PubMed]
109. Kim, J.E.; Park, Y.J. Improved antitumor efficacy of hyaluronic acid-complexed paclitaxel nanoemulsions in
treating non-small cell lung cancer. Biomol. Ther. (Seoul) 2017, 25, 411–416. [CrossRef] [PubMed]
110. Najlah, M.; Kadam, A.; Wan, K.W.; Ahmed, W.; Taylor, K.M.; Elhissi, A.M. Novel paclitaxel formulations
solubilized by parenteral nutrition nanoemulsions for application against glioma cell lines. Int. J. Pharm.
2016, 506, 102–109. [CrossRef] [PubMed]
111. Zhang, L.; Hayes, D.G.; Chen, G.; Zhong, Q. Transparent dispersions of milk-fat-based nanostructured lipid
carriers for delivery of beta-carotene. J. Agric. Food Chem. 2013, 61, 9435–9443. [CrossRef] [PubMed]
112. Zhao, X.L.; Yang, C.R.; Yang, K.L.; Li, K.X.; Hu, H.Y.; Chen, D.W. Preparation and characterization of
nanostructured lipid carriers loaded traditional Chinese medicine, zedoary turmeric oil. Drug Dev. Ind. Pharm.
2010, 36, 773–780. [CrossRef] [PubMed]
113. Mehmood, T.; Ahmad, A.; Ahmed, A.; Ahmed, Z. Optimization of olive oil based O/W nanoemulsions
prepared through ultrasonic homogenization: A response surface methodology approach. Food Chem. 2017,
229, 790–796. [CrossRef] [PubMed]
114. Koutelidakis, A.E.; Argyri, K.; Sevastou, Z.; Lamprinaki, D.; Panagopoulou, E.; Paximada, E.; Sali, A.;
Papalazarou, V.; Mallouchos, A.; Evageliou, V.; et al. Bioactivity of epigallocatechin gallate nanoemulsions
evaluated in mice model. J. Med. Food 2017, 20, 923–931. [CrossRef] [PubMed]
115. Rodriguez-Burneo, N.; Busquets, M.A.; Estelrich, J. Magnetic Nanoemulsions: Comparison between
nanoemulsions formed by ultrasonication and by spontaneous emulsification. Nanomaterials 2017, 7.
[CrossRef] [PubMed]
116. Abdelwahab, S.I.; Sheikh, B.Y.; Taha, M.M.; How, C.W.; Abdullah, R.; Yagoub, U.; El-Sunousi, R.; Eid, E.E.
Thymoquinone-loaded nanostructured lipid carriers: Preparation, gastroprotection, in vitro toxicity, and
pharmacokinetic properties after extravascular administration. Int. J. Nanomed. 2013, 8, 2163–2172. [CrossRef]
[PubMed]
117. Mikulcova, V.; Kasparkova, V.; Humpolicek, P.; Bunkova, L. Formulation, characterization and properties of
hemp seed oil and its emulsions. Molecules 2017, 22, E700. [CrossRef] [PubMed]
118. Lu, R.; Liu, S.; Wang, Q.; Li, X. Nanoemulsions as novel oral carriers of stiripentol: Insights into the protective
effect and absorption enhancement. Int. J. Nanomed. 2015, 10, 4937–4946.
119. Panatieri, L.F.; Brazil, N.T.; Faber, K.; Medeiros-Neves, B.; von Poser, G.L.; Rott, M.B.; Zorzi, G.K.; Teixeira, H.F.
Nanoemulsions containing a coumarin-rich extract from pterocaulon balansae (Asteraceae) for the treatment
of ocular acanthamoeba keratitis. AAPS PharmSciTech 2017, 18, 721–728. [CrossRef] [PubMed]
120. Ragelle, H.; Crauste-Manciet, S.; Seguin, J.; Brossard, D.; Scherman, D.; Arnaud, P.; Chabot, G.G.
Nanoemulsion formulation of fisetin improves bioavailability and antitumour activity in mice. Int. J. Pharm.
2012, 427, 452–459. [CrossRef] [PubMed]
121. Hejri, A.; Khosravi, A.; Gharanjig, K.; Hejazi, M. Optimisation of the formulation of beta-carotene loaded
nanostructured lipid carriers prepared by solvent diffusion method. Food Chem. 2013, 141, 117–123.
[CrossRef] [PubMed]
122. Tan, S.W.; Billa, N.; Roberts, C.R.; Burley, J.C. Surfactant effects on the physical characteristics of Amphotericin
B-containing nanostructured lipid carriers. Colloids Surf. Physicochem. Eng. Aspects 2010, 372, 73–79.
[CrossRef]
123. Wang, L.; Luo, Q.; Lin, T.; Li, R.; Zhu, T.; Zhou, K.; Ji, Z.; Song, J.; Jia, B.; Zhang, C.; et al. PEGylated
nanostructured lipid carriers (PEG-NLC) as a novel drug delivery system for biochanin A. Drug Dev.
Ind. Pharm. 2015, 41, 1204–1212. [CrossRef] [PubMed]
124. Sood, J.; Sapra, B.; Tiwary, A.K. Microemulsion transdermal formulation for simultaneous delivery of
valsartan and nifedipine: Formulation by design. AAPS PharmSciTech 2017, 18, 1901–1916. [CrossRef]
[PubMed]
125. Dash, R.N.; Mohammed, H.; Humaira, T.; Ramesh, D. Design, optimization and evaluation of glipizide
solid self-nanoemulsifying drug delivery for enhanced solubility and dissolution. Saudi Pharm. J. 2015,
23, 528–540. [CrossRef] [PubMed]
126. Bondi, M.L.; Botto, C.; Amore, E.; Emma, M.R.; Augello, G.; Craparo, E.F.; Cervello, M. Lipid nanocarriers
containing sorafenib inhibit colonies formation in human hepatocarcinoma cells. Int. J. Pharm. 2015,
493, 75–85. [CrossRef] [PubMed]
Pharmaceutics 2018, 10, 74 28 of 33
127. Sugumaran, A.; Ponnusamy, C.; Kandasamy, P.; Krishnaswami, V.; Palanichamy, R.; Kandasamy, R.;
Lakshmanan, M.; Natesan, S. Development and evaluation of camptothecin loaded polymer stabilized
nanoemulsion: Targeting potential in 4T1-breast tumour xenograft model. Eur. J. Pharm. Sci. 2018, 116, 15–25.
[CrossRef] [PubMed]
128. Griesser, J.; Burtscher, S.; Kollner, S.; Nardin, I.; Prufert, F.; Bernkop-Schnurch, A. Zeta potential changing
self-emulsifying drug delivery systems containing phosphorylated polysaccharides. Eur. J. Pharm. Biopharm.
2017, 119, 264–270. [CrossRef] [PubMed]
129. Shah, A.V.; Desai, H.H.; Thool, P.; Dalrymple, D.; Serajuddin, A.T.M. Development of self-microemulsifying
drug delivery system for oral delivery of poorly water-soluble nutraceuticals. Drug Dev. Ind. Pharm. 2018,
44, 895–901. [CrossRef] [PubMed]
130. Zhang, X.; Pan, W.; Gan, L.; Zhu, C.; Gan, Y.; Nie, S. Preparation of a dispersible PEGylate nanostructured
lipid carriers (NLC) loaded with 10-hydroxycamptothecin by spray-drying. Chem. Pharm. Bull. 2008,
56, 1645–1650. [CrossRef] [PubMed]
131. Kaur, A.; Katiyar, S.S.; Kushwah, V.; Jain, S. Nanoemulsion loaded gel for topical co-delivery of clobitasol
propionate and calcipotriol in psoriasis. Nanomedicine 2017, 13, 1473–1482. [CrossRef] [PubMed]
132. Patel, G.; Shelat, P.; Lalwani, A. Statistical modeling, optimization and characterization of solid
self-nanoemulsifying drug delivery system of lopinavir using design of experiment. Drug Deliv. 2016,
23, 3027–3042. [CrossRef] [PubMed]
133. Shah, N.V.; Seth, A.K.; Balaraman, R.; Aundhia, C.J.; Maheshwari, R.A.; Parmar, G.R. Nanostructured lipid
carriers for oral bioavailability enhancement of raloxifene: Design and in vivo study. J. Adv. Res. 2016,
7, 423–434. [CrossRef] [PubMed]
134. Elnaggar, Y.S.; El-Massik, M.A.; Abdallah, O.Y. Self-nanoemulsifying drug delivery systems of tamoxifen
citrate: Design and optimization. Int. J. Pharm. 2009, 380, 133–141. [CrossRef] [PubMed]
135. Bandyopadhyay, S.; Katare, O.P.; Singh, B. Optimized self nano-emulsifying systems of ezetimibe with
enhanced bioavailability potential using long chain and medium chain triglycerides. Colloids Surf.
B Biointerfaces 2012, 100, 50–61. [CrossRef] [PubMed]
136. Kamboj, S.; Rana, V. Quality-by-design based development of a self-microemulsifying drug delivery system
to reduce the effect of food on Nelfinavir mesylate. Int. J. Pharm. 2016, 501, 311–325. [CrossRef] [PubMed]
137. Shi, F.; Yang, G.; Ren, J.; Guo, T.; Du, Y.; Feng, N. Formulation design, preparation, and in vitro and in vivo
characterizations of beta-Elemene-loaded nanostructured lipid carriers. Int. J. Nanomed. 2013, 8, 2533–2541.
[CrossRef] [PubMed]
138. Mustapha, O.; Kim, K.S.; Shafique, S.; Kim, D.S.; Jin, S.G.; Seo, Y.G.; Youn, Y.S.; Oh, K.T.; Lee, B.J.; Park, Y.J.;
et al. Development of novel cilostazol-loaded solid SNEDDS using a SPG membrane emulsification
technique: Physicochemical characterization and in vivo evaluation. Colloids Surf. B Biointerfaces 2017,
150, 216–222. [CrossRef] [PubMed]
139. Esposito, E.; Drechsler, M.; Mariani, P.; Carducci, F.; Servadio, M.; Melancia, F.; Ratano, P.; Campolongo, P.;
Trezza, V.; Cortesi, R.; et al. Lipid nanoparticles for administration of poorly water soluble neuroactive drugs.
Biomed. Microdevices 2017, 19, 44. [CrossRef] [PubMed]
140. Dawoud, M.Z.; Nasr, M. Comparison of drug release from liquid crystalline monoolein dispersions and
solid lipid nanoparticles using a flow cytometric technique. Acta Pharm. Sin. B 2016, 6, 163–169. [CrossRef]
[PubMed]
141. Brusewitz, C.; Schendler, A.; Funke, A.; Wagner, T.; Lipp, R. Novel poloxamer-based nanoemulsions to
enhance the intestinal absorption of active compounds. Int. J. Pharm. 2007, 329, 173–181. [CrossRef]
[PubMed]
142. Seo, Y.G.; Kim, D.W.; Cho, K.H.; Yousaf, A.M.; Kim, D.S.; Kim, J.H.; Kim, J.O.; Yong, C.S.; Choi, H.G.
Preparation and pharmaceutical evaluation of new tacrolimus-loaded solid self-emulsifying drug delivery
system. Arch. Pharm. Res. 2015, 38, 223–228. [CrossRef] [PubMed]
143. Thatipamula, R.; Palem, C.; Gannu, R.; Mudragada, S.; Yamsani, M. Formulation and in vitro characterization
of domperidone loaded solid lipid nanoparticles and nanostructured lipid carriers. Daru 2011, 19, 23–32.
[PubMed]
144. Hsu, H.J.; Huang, R.F.; Kao, T.H.; Inbaraj, B.S.; Chen, B.H. Preparation of carotenoid extracts and
nanoemulsions from Lycium barbarum L. and their effects on growth of HT-29 colon cancer cells.
Nanotechnology 2017, 28, 135103. [CrossRef] [PubMed]
Pharmaceutics 2018, 10, 74 29 of 33
145. Chavan, R.B.; Modi, S.R.; Bansal, A.K. Role of solid carriers in pharmaceutical performance of solid
supersaturable SEDDS of celecoxib. Int. J. Pharm. 2015, 495, 374–384. [CrossRef] [PubMed]
146. Elmowafy, M.; Ibrahim, H.M.; Ahmed, M.A.; Shalaby, K.; Salama, A.; Hefesha, H. Atorvastatin-loaded
nanostructured lipid carriers (NLCs): Strategy to overcome oral delivery drawbacks. Drug Deliv. 2017,
24, 932–941. [CrossRef] [PubMed]
147. Hong, L.; Zhou, C.L.; Chen, F.P.; Han, D.; Wang, C.Y.; Li, J.X.; Chi, Z.; Liu, C.G. Development of a
carboxymethyl chitosan functionalized nanoemulsion formulation for increasing aqueous solubility, stability
and skin permeability of astaxanthin using low-energy method. J. Microencapsul. 2017, 34, 707–721.
[CrossRef] [PubMed]
148. Kamble, R.N.; Mehta, P.P.; Kumar, A. Efavirenz self-nano-emulsifying drug delivery system: In vitro and
in vivo evaluation. AAPS PharmSciTech 2016, 17, 1240–1247. [CrossRef] [PubMed]
149. Safwat, S.; Ishak, R.A.H.; Hathout, R.M.; Mortada, N.D. Nanostructured lipid carriers loaded with
simvastatin: Effect of PEG/glycerides on characterization, stability, cellular uptake efficiency and in vitro
cytotoxicity. Drug Dev. Ind. Pharm. 2017, 43, 1112–1125. [CrossRef] [PubMed]
150. Cuomo, F.; Cofelice, M.; Venditti, F.; Ceglie, A.; Miguel, M.; Lindman, B.; Lopez, F. In-vitro digestion of
curcumin loaded chitosan-coated liposomes. Colloids Surf. B. Biointerfaces 2018, 168, 29–34. [CrossRef]
[PubMed]
151. Freag, M.S.; Saleh, W.M.; Abdallah, O.Y. Self-assembled phospholipid-based phytosomal nanocarriers as
promising platforms for improving oral bioavailability of the anticancer celastrol. Int. J. Pharm. 2017,
535, 18–26. [CrossRef] [PubMed]
152. Zhang, X.; Zhang, T.; Zhou, X.; Liu, H.; Sun, H.; Ma, Z.; Wu, B. Enhancement of oral bioavailability of
tripterine through lipid nanospheres: Preparation, characterization, and absorption evaluation. J. Pharm. Sci.
2014, 103, 1711–1719. [CrossRef] [PubMed]
153. Wu, B.; Lu, S.T.; Zhang, L.J.; Zhuo, R.X.; Xu, H.B.; Huang, S.W. Codelivery of doxorubicin and triptolide with
reduction-sensitive lipid-polymer hybrid nanoparticles for in vitro and in vivo synergistic cancer treatment.
Int. J. Nanomed. 2017, 12, 1853–1862. [CrossRef] [PubMed]
154. Yao, C.; Wu, M.; Zhang, C.; Lin, X.; Wei, Z.; Zheng, Y.; Da, Z.; Zhang, Z.; Liu, X. Photoresponsive
lipid-polymer hybrid nanoparticles for controlled doxorubicin release. Nanotechnology 2017, 28, 255101.
[CrossRef] [PubMed]
155. Valicherla, G.R.; Dave, K.M.; Syed, A.A.; Riyazuddin, M.; Gupta, A.P.; Singh, A.; Mitra, K.; Datta, D.;
Gayen, J.R. Formulation optimization of Docetaxel loaded self-emulsifying drug delivery system to enhance
bioavailability and anti-tumor activity. Sci. Rep. 2016, 6, 26895. [CrossRef] [PubMed]
156. Gaber, D.M.; Nafee, N.; Abdallah, O.Y. Myricetin solid lipid nanoparticles: Stability assurance from system
preparation to site of action. Eur. J. Pharm. Sci. 2017, 109, 569–580. [CrossRef] [PubMed]
157. Joseph, E.; Reddi, S.; Rinwa, V.; Balwani, G.; Saha, R. Design and in vivo evaluation of solid lipid
nanoparticulate systems of Olanzapine for acute phase schizophrenia treatment: Investigations on
antipsychotic potential and adverse effects. Eur. J. Pharm. Sci. 2017, 104, 315–325. [CrossRef] [PubMed]
158. Goncalves, L.M.; Maestrelli, F.; Di Cesare Mannelli, L.; Ghelardini, C.; Almeida, A.J.; Mura, P. Development
of solid lipid nanoparticles as carriers for improving oral bioavailability of glibenclamide. Eur. J. Pharm.
Biopharm. 2016, 102, 41–50. [CrossRef] [PubMed]
159. Yu, Q.; Hu, X.; Ma, Y.; Xie, Y.; Lu, Y.; Qi, J.; Xiang, L.; Li, F.; Wu, W. Lipids-based nanostructured lipid carriers
(NLCs) for improved oral bioavailability of sirolimus. Drug Deliv. 2016, 23, 1469–1475. [CrossRef] [PubMed]
160. Makwana, V.; Jain, R.; Patel, K.; Nivsarkar, M.; Joshi, A. Solid lipid nanoparticles (SLN) of Efavirenz as
lymph targeting drug delivery system: Elucidation of mechanism of uptake using chylomicron flow blocking
approach. Int. J. Pharm. 2015, 495, 439–446. [CrossRef] [PubMed]
161. Elmowafy, M.; Samy, A.; Raslan, M.A.; Salama, A.; Said, R.A.; Abdelaziz, A.E.; El-Eraky, W.; El Awdan, S.;
Viitala, T. Enhancement of bioavailability and pharmacodynamic effects of thymoquinone via nanostructured
lipid carrier (NLC) formulation. AAPS PharmSciTech 2016, 17, 663–672. [CrossRef] [PubMed]
162. Zhang, S.; Meng, X.; Wang, Z.; Fan, A.; Wang, G.; Zhao, Y.; Tang, Y. Engineering hot-melt extruded solid
dispersion for controlled release of hydrophilic drugs. Eur. J. Pharm. Sci. 2017, 100, 109–115. [CrossRef]
[PubMed]
Pharmaceutics 2018, 10, 74 30 of 33
163. Xu, W.; Lim, S.J.; Lee, M.K. Cellular uptake and antitumour activity of paclitaxel incorporated into
trilaurin-based solid lipid nanoparticles in ovarian cancer. J. Microencapsul. 2013, 30, 755–761. [CrossRef]
[PubMed]
164. Christophersen, P.C.; Zhang, L.; Mullertz, A.; Nielsen, H.M.; Yang, M.; Mu, H. Solid lipid particles for oral
delivery of peptide and protein drugs II–the digestion of trilaurin protects desmopressin from proteolytic
degradation. Pharm. Res. 2014, 31, 2420–2428. [CrossRef] [PubMed]
165. Yao, M.; McClements, D.J.; Xiao, H. Improving oral bioavailability of nutraceuticals by engineered
nanoparticle-based delivery systems. Curr. Opin. Food Sci. 2015, 2, 14–19. [CrossRef]
166. Keck, C.M.; Kovacevic, A.; Muller, R.H.; Savic, S.; Vuleta, G.; Milic, J. Formulation of solid lipid nanoparticles
(SLN): The value of different alkyl polyglucoside surfactants. Int. J. Pharm. 2014, 474, 33–41. [CrossRef]
[PubMed]
167. Ribeiro, L.N.; Franz-Montan, M.; Breitkreitz, M.C.; Alcantara, A.C.; Castro, S.R.; Guilherme, V.A.;
Barbosa, R.M.; de Paula, E. Nanostructured lipid carriers as robust systems for topical lidocaine-prilocaine
release in dentistry. Eur. J. Pharm. Sci. 2016, 93, 192–202. [CrossRef] [PubMed]
168. Vijayakumar, A.; Baskaran, R.; Jang, Y.S.; Oh, S.H.; Yoo, B.K. Quercetin-loaded solid lipid nanoparticle
dispersion with improved physicochemical properties and cellular uptake. AAPS PharmSciTech 2017,
18, 875–883. [CrossRef] [PubMed]
169. Tsai, M.J.; Wu, P.C.; Huang, Y.B.; Chang, J.S.; Lin, C.L.; Tsai, Y.H.; Fang, J.Y. Baicalein loaded in tocol
nanostructured lipid carriers (tocol NLCs) for enhanced stability and brain targeting. Int. J. Pharm. 2012,
423, 461–470. [CrossRef] [PubMed]
170. Feeney, O.M.; Crum, M.F.; McEvoy, C.L.; Trevaskis, N.L.; Williams, H.D.; Pouton, C.W.; Charman, W.N.;
Bergstrom, C.A.S.; Porter, C.J.H. 50 years of oral lipid-based formulations: Provenance, progress and future
perspectives. Adv. Drug Deliv. Rev. 2016, 101, 167–194. [CrossRef] [PubMed]
171. Taylor, L.S.; Zhang, G.G.Z. Physical chemistry of supersaturated solutions and implications for oral
absorption. Adv. Drug Deliv. Rev. 2016, 101, 122–142. [CrossRef] [PubMed]
172. Shekhawat, P.B.; Pokharkar, V.B. Understanding peroral absorption: Regulatory aspects and contemporary
approaches to tackling solubility and permeability hurdles. Acta Pharm. Sin. B 2017, 7, 260–280. [CrossRef]
[PubMed]
173. Wang, H.; Wang, H.; Yang, W.; Yu, M.; Sun, S.; Xie, B. Improved oral bioavailability and liver targeting of
sorafenib solid lipid nanoparticles in rats. AAPS PharmSciTech 2018, 19, 761–768. [CrossRef] [PubMed]
174. Zhou, X.; Zhang, X.; Ye, Y.; Zhang, T.; Wang, H.; Ma, Z.; Wu, B. Nanostructured lipid carriers used for
oral delivery of oridonin: An effect of ligand modification on absorption. Int. J. Pharm. 2015, 479, 391–398.
[CrossRef] [PubMed]
175. Li, W.; Zhang, T.; Ye, Y.; Zhang, X.; Wu, B. Enhanced bioavailability of tripterine through lipid nanoparticles
using broccoli-derived lipids as a carrier material. Int. J. Pharm. 2015, 495, 948–955. [CrossRef] [PubMed]
176. Chavez-Zamudio, R.; Ochoa-Flores, A.A.; Soto-Rodriguez, I.; Garcia-Varela, R.; Garcia, H.S. Preparation,
characterization and bioavailability by oral administration of O/W curcumin nanoemulsions stabilized with
lysophosphatidylcholine. Food Funct. 2017, 8, 3346–3354. [CrossRef] [PubMed]
177. Ge, L.; He, X.; Zhang, Y.; Zhang, Y.; Chai, F.; Jiang, L.; Webster, T.J.; Zheng, C. A dabigatran etexilate
phospholipid complex nanoemulsion system for further oral bioavailability by reducing drug-leakage in the
gastrointestinal tract. Nanomedicine 2017. [CrossRef] [PubMed]
178. Rushmi, Z.T.; Akter, N.; Mow, R.J.; Afroz, M.; Kazi, M.; de Matas, M.; Rahman, M.; Shariare, M.H. The impact
of formulation attributes and process parameters on black seed oil loaded liposomes and their performance
in animal models of analgesia. Saudi Pharm. J. 2017, 25, 404–412. [CrossRef] [PubMed]
179. Hu, X.; Zhang, J.; Yu, Z.; Xie, Y.; He, H.; Qi, J.; Dong, X.; Lu, Y.; Zhao, W.; Wu, W. Environment-responsive
aza-BODIPY dyes quenching in water as potential probes to visualize the in vivo fate of lipid-based
nanocarriers. Nanomedicine 2015, 11, 1939–1948. [CrossRef] [PubMed]
180. Xia, F.; Fan, W.; Jiang, S.; Ma, Y.; Lu, Y.; Qi, J.; Ahmad, E.; Dong, X.; Zhao, W.; Wu, W. Size-dependent
translocation of nanoemulsions via oral delivery. ACS Appl. Mater. Interfaces 2017, 9, 21660–21672. [CrossRef]
[PubMed]
181. Heurtault, B.; Saulnier, P.; Pech, B.; Proust, J.E.; Benoit, J.P. Physico-chemical stability of colloidal lipid
particles. Biomaterials 2003, 24, 4283–4300. [CrossRef]
Pharmaceutics 2018, 10, 74 31 of 33
182. Tan, A.; Rao, S.; Prestidge, C.A. Transforming lipid-based oral drug delivery systems into solid dosage forms:
An overview of solid carriers, physicochemical properties, and biopharmaceutical performance. Pharm. Res.
2013, 30, 2993–3017. [CrossRef] [PubMed]
183. Howard, M.D.; Lu, X.; Jay, M.; Dziubla, T.D. Optimization of the lyophilization process for long-term stability
of solid-lipid nanoparticles. Drug Dev. Ind. Pharm. 2012, 38, 1270–1279. [CrossRef] [PubMed]
184. Freag, M.S.; Elnaggar, Y.S.; Abdallah, O.Y. Lyophilized phytosomal nanocarriers as platforms for enhanced
diosmin delivery: Optimization and ex vivo permeation. Int. J. Nanomed. 2013, 8, 2385–2397.
185. Wang, T.; Hu, Q.; Zhou, M.; Xue, J.; Luo, Y. Preparation of ultra-fine powders from polysaccharide-coated
solid lipid nanoparticles and nanostructured lipid carriers by innovative nano spray drying technology.
Int. J. Pharm. 2016, 511, 219–222. [CrossRef] [PubMed]
186. Wang, T.; Hu, Q.; Zhou, M.; Xia, Y.; Nieh, M.P.; Luo, Y. Development of “all natural” layer-by-layer
redispersible solid lipid nanoparticles by nano spray drying technology. Eur. J. Pharm. Biopharm. 2016,
107, 273–285. [CrossRef] [PubMed]
187. Tian, Z.; Yi, Y.; Yuan, H.; Han, J.; Zhang, X.; Xie, Y.; Lu, Y.; Qi, J.; Wu, W. Solidification of nanostructured lipid
carriers (NLCs) onto pellets by fluid-bed coating: Preparation, in vitro characterization and bioavailability
in dogs. Powder Technol. 2013, 247, 120–127. [CrossRef]
188. Ali, M.E.; Lamprecht, A. Spray freeze drying as an alternative technique for lyophilization of polymeric and
lipid-based nanoparticles. Int. J. Pharm. 2017, 516, 170–177. [CrossRef] [PubMed]
189. Chouhan, N.; Mittal, V.; Kaushik, D.; Khatkar, A.; Raina, M. Self emulsifying drug delivery system (SEDDS)
for phytoconstituents: A review. Curr. Drug Deliv. 2015, 12, 244–253. [CrossRef] [PubMed]
190. Keohane, K.; Rosa, M.; Coulter, I.S.; Griffin, B.T. Enhanced colonic delivery of ciclosporin A self-emulsifying
drug delivery system encapsulated in coated minispheres. Drug Dev. Ind. Pharm. 2016, 42, 245–253.
[CrossRef] [PubMed]
191. Patel, J.; Dhingani, A.; Garala, K.; Raval, M.; Sheth, N. Quality by design approach for oral bioavailability
enhancement of irbesartan by self-nanoemulsifying tablets. Drug Deliv. 2014, 21, 412–435. [CrossRef]
[PubMed]
192. Nnamani, P.O.; Ugwu, A.A.; Ibezim, E.C.; Kenechukwu, F.C.; Akpa, P.A.; Ogbonna, J.N.; Obitte, N.C.;
Odo, A.N.; Windbergs, M.; Lehr, C.M.; et al. Sustained-release liquisolid compact tablets containing
artemether-lumefantrine as alternate-day regimen for malaria treatment to improve patient compliance.
Int. J. Nanomed. 2016, 11, 6365–6378. [CrossRef] [PubMed]
193. Spireas, S.; Sadu, S.; Grover, R. In vitro release evaluation of hydrocortisone liquisolid tablets. J. Pharm. Sci.
1998, 87, 867–872. [CrossRef] [PubMed]
194. Nokhodchi, A.; Hentzschel, C.M.; Leopold, C.S. Drug release from liquisolid systems: Speed it up, slow it
down. Expert Opin. Drug Deliv. 2011, 8, 191–205. [CrossRef] [PubMed]
195. Kamble, P.R.; Shaikh, K.S.; Chaudhari, P.D. Application of liquisolid technology for enhancing solubility and
dissolution of rosuvastatin. Adv. Pharm. Bull. 2014, 4, 197–204. [PubMed]
196. Badawy, M.A.; Kamel, A.O.; Sammour, O.A. Use of biorelevant media for assessment of a poorly soluble
weakly basic drug in the form of liquisolid compacts: In vitro and in vivo study. Drug Deliv. 2016, 23, 818–827.
[PubMed]
197. Khames, A. Liquisolid technique: A promising alternative to conventional coating for improvement of drug
photostability in solid dosage forms. Expert Opin. Drug Deliv. 2013, 10, 1335–1343. [CrossRef] [PubMed]
198. Sharma, V.; Pathak, K. Effect of hydrogen bond formation/replacement on solubility characteristics, gastric
permeation and pharmacokinetics of curcumin by application of powder solution technology. Acta Pharm.
Sin. B 2016, 6, 600–613. [CrossRef] [PubMed]
199. Yehia, S.A.; El-Ridi, M.S.; Tadros, M.I.; El-Sherif, N.G. Enhancement of the oral bioavailability of fexofenadine
hydrochloride via Cremophor® El-based liquisolid tablets. Adv. Pharm. Bull. 2015, 5, 569–581. [CrossRef]
[PubMed]
200. Chella, N.; Narra, N.; Rama Rao, T. Preparation and characterization of liquisolid compacts for improved
dissolution of telmisartan. J. Drug Deliv. 2014. [CrossRef] [PubMed]
201. Elkordy, A.A.; Essa, E.A.; Dhuppad, S.; Jammigumpula, P. Liquisolid technique to enhance and to sustain
griseofulvin dissolution: Effect of choice of non-volatile liquid vehicles. Int. J. Pharm. 2012, 434, 122–132.
[CrossRef] [PubMed]
Pharmaceutics 2018, 10, 74 32 of 33
202. Khames, A. Investigation of the effect of solubility increase at the main absorption site on bioavailability
of BCS class II drug (risperidone) using liquisolid technique. Drug Deliv. 2017, 24, 328–338. [CrossRef]
[PubMed]
203. Javadzadeh, Y.; Jafari-Navimipour, B.; Nokhodchi, A. Liquisolid technique for dissolution rate enhancement
of a high dose water-insoluble drug (carbamazepine). Int. J. Pharm. 2007, 341, 26–34. [CrossRef] [PubMed]
204. Kala, N.P.; Shastri, D.H.; Shelat, P.K. Design and Characterization of buccoadhesive liquisolid system of an
antihypertensive drug. J. Drug Deliv. 2015, 2015, 574247. [CrossRef] [PubMed]
205. Hentzschel, C.M.; Alnaief, M.; Smirnova, I.; Sakmann, A.; Leopold, C.S. Enhancement of griseofulvin release
from liquisolid compacts. Eur. J. Pharm. Biopharm. 2012, 80, 130–135. [CrossRef] [PubMed]
206. Vranikova, B.; Gajdziok, J. Evaluation of sorptive properties of various carriers and coating materials for
liquisolid systems. Acta Pol. Pharm. 2015, 72, 539–549. [PubMed]
207. Prajapati, S.T.; Bulchandani, H.H.; Patel, D.M.; Dumaniya, S.K.; Patel, C.N. Formulation and evaluation of
liquisolid compacts for olmesartan medoxomil. J. Drug Deliv. 2013. [CrossRef] [PubMed]
208. Venkateswarlu, K.; Preethi, J.K.; Chandrasekhar, K.B. Enhancement of loperamide dissolution rate by
liquisolid compact technique. Adv. Pharm. Bull. 2016, 6, 385–390. [CrossRef] [PubMed]
209. Hentzschel, C.M.; Sakmann, A.; Leopold, C.S. Suitability of various excipients as carrier and coating materials
for liquisolid compacts. Drug Dev. Ind. Pharm. 2011, 37, 1200–1207. [CrossRef] [PubMed]
210. Lu, M.; Xing, H.; Jiang, J.; Chen, X.; Yang, T.; Wang, D.; Ding, P. Liquisolid technique and its applications in
pharmaceutics. Asian J. Pharm. Sci. 2017, 12, 115–123. [CrossRef]
211. Javadzadeh, Y.; Musaalrezaei, L.; Nokhodchi, A. Liquisolid technique as a new approach to sustain
propranolol hydrochloride release from tablet matrices. Int. J. Pharm. 2008, 362, 102–108. [CrossRef]
[PubMed]
212. Karmarkar, A.B.; Gonjari, I.D.; Hosmani, A.H. Liquisolid technology for dissolution rate enhancement or
sustained release. Expert Opin. Drug Deliv. 2010, 7, 1227–1234. [CrossRef] [PubMed]
213. Karmarkar, A.B.; Gonjari, I.D.; Hosmani, A.H.; Dhabale, P.N. Evaluation of in vitro dissolution profile
comparison methods of sustained release tramadol hydrochloride liquisolid compact formulations with
marketed sustained release tablets. Drug Discov. Ther. 2010, 4, 26–32. [PubMed]
214. Spireas, S.S.; Jarowski, C.I.; Rohera, B.D. Powdered solution technology: Principles and mechanism.
Pharm. Res. 1992, 9, 1351–1358. [CrossRef] [PubMed]
215. Kulkarni, A.S.; Aloorkar, N.H.; Mane, M.S.; Gaja, J.B. Liquisolid systems: A review. Int. J. Pharm. Sci. Nanotechnol.
2010, 3, 795–802.
216. Gavali, S.M.; Pacharane, S.S.; Sankpal, S.V.; Jadhav, K.R.; Kadam, V.J. Liquisolid compact: A new technique
for enhancement of drug dissolution. Int. J. Res. Pharm. Chem. 2011, 1, 705–713.
217. Barmpalexis, P.; Grypioti, A.; Eleftheriadis, G.K.; Fatouros, D.G. Development of a new aprepitant liquisolid
formulation with the aid of artificial neural networks and genetic programming. AAPS PharmSciTech 2018,
19, 741–752. [CrossRef] [PubMed]
218. Karmarkar, A.B. Effect of Ceolus KG-802 on the dissolution rate of fenofibrate liquisolid tablets:
Preformulation and formulation development studies. Drug Discov. Ther. 2010, 4, 493–498. [PubMed]
219. Saeedi, M.; Akbari, J.; Morteza-Semnani, K.; Enayati-Fard, R.; Sar-Reshteh-Dar, S.; Soleymani, A.
Enhancement of dissolution rate of indomethacin: Using liquisolid compacts. Iran. J. Pharm. Sci. 2011,
10, 25–34.
220. Vittal, G.V.; Deveswaran, R.; Bharath, S.; Basavaraj, B.; Madhavan, V. Formulation and characterization of
ketoprofen liquisolid compacts by Box-Behnken design. Int. J. Pharm. Investig. 2012, 2, 150–156. [PubMed]
221. El-Hammadi, M.; Awad, N. Investigating the use of liquisolid compacts technique to minimize the influence
of pH variations on loratadine release. AAPS PharmSciTech 2012, 13, 53–58. [CrossRef] [PubMed]
222. Vaskula, S.; Vemula, S.K.; Bontha, V.K.; Garrepally, P. Liquisolid compacts: An approach to enhance the
dissolution rate of nimesulide. J. Appl. Pharm. Sci. 2012, 2, 115–121.
223. Akinlade, B.; Elkordy, A.A.; Essa, E.A.; Elhagar, S. Liquisolid systems to improve the dissolution of
furosemide. Sci. Pharm. 2010, 78, 325–344. [CrossRef] [PubMed]
224. Patel, D.S.; Pipaliya, R.M.; Surti, N. Liquisolid tablets for dissolution enhancement of a hypolipidemic drug.
Indian J. Pharm. Sci. 2015, 77, 290–298. [PubMed]
225. Chella, N.; Shastri, N.; Tadikonda, R.R. Use of the liquisolid compact technique for improvement of the
dissolution rate of valsartan. Acta Pharm. Sin. B 2012, 2, 502–508. [CrossRef]
Pharmaceutics 2018, 10, 74 33 of 33
226. Shin, S. Dissolution characteristics of furosemide-polymer coprecipitates. Arch. Pharm. Res. 1979, 2, 35–48.
[CrossRef]
227. Shin, S.-C.; Oh, I.-J.; Lee, Y.-B.; Choi, H.-K.; Choi, J.-S. Enhanced dissolution of furosemide by coprecipitating
or cogrinding with crospovidone. Int. J. Pharm. 1998, 175, 17–24. [CrossRef]
228. Planinsek, O.; Kovacic, B.; Vrecer, F. Carvedilol dissolution improvement by preparation of solid dispersions
with porous silica. Int. J. Pharm. 2011, 406, 41–48. [CrossRef] [PubMed]
229. Karagianni, A.; Malamatari, M.; Kachrimanis, K. Pharmaceutical cocrystals: New solid phase modification
approaches for the formulation of APIs. Pharmaceutics 2018, 10, E18. [CrossRef] [PubMed]
230. Thakuria, R.; Delori, A.; Jones, W.; Lipert, M.P.; Roy, L.; Rodriguez-Hornedo, N. Pharmaceutical cocrystals
and poorly soluble drugs. Int. J. Pharm. 2013, 453, 101–125. [CrossRef] [PubMed]
231. Du, S.; Wang, Y.; Wu, S.; Yu, B.; Shi, P.; Bian, L.; Zhang, D.; Hou, J.; Wang, J.; Gong, J. Two novel cocrystals of
lamotrigine with isomeric bipyridines and in situ monitoring of the cocrystallization. Eur. J. Pharm. Sci. 2017,
110, 19–25. [CrossRef] [PubMed]
232. Yamashita, H.; Sun, C.C. Improving dissolution rate of carbamazepine-glutaric acid cocrystal through
solubilization by excess coformer. Pharm. Res. 2017, 35, 4. [CrossRef] [PubMed]
233. Semalty, A. Cyclodextrin and phospholipid complexation in solubility and dissolution enhancement:
A critical and meta-analysis. Expert Opin. Drug Deliv. 2014, 11, 1255–1272. [CrossRef] [PubMed]
234. Mady, F.M.; Farghaly Aly, U. Experimental, molecular docking investigations and bioavailability study
on the inclusion complexes of finasteride and cyclodextrins. Drug Des. Dev. Ther. 2017, 11, 1681–1692.
[CrossRef] [PubMed]
235. Loftsson, T.; Moya-Ortega, M.D.; Alvarez-Lorenzo, C.; Concheiro, A. Pharmacokinetics of cyclodextrins and
drugs after oral and parenteral administration of drug/cyclodextrin complexes. J. Pharm. Pharmacol. 2016,
68, 544–555. [CrossRef] [PubMed]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).