Sunteți pe pagina 1din 15

European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813

Contents lists available at ScienceDirect

European Journal of Pharmaceutics and Biopharmaceutics


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

Review article

Current trends and future perspectives of solid dispersions containing


poorly water-soluble drugs
Chau Le-Ngoc Vo, Chulhun Park, Beom-Jin Lee ⇑
Bioavailability Control Laboratory, College of Pharmacy, Ajou University, Suwon, Republic of Korea

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

Article history: Over 40% of active pharmaceutical ingredients (API) in development pipelines are poorly water-soluble
Received 8 April 2013 drugs which limit formulation approaches, clinical application and marketability because of their low
Accepted in revised form 9 September 2013 dissolution and bioavailability. Solid dispersion has been considered one of the major advancements in
Available online 18 September 2013
overcoming these issues with several successfully marketed products. A number of key references that
describe state-of-the-art technologies have been collected in this review, which addresses various
Keywords: pharmaceutical strategies and future visions for the solubilization of poorly water-soluble drugs accord-
Classification of solid dispersions
ing to the four generations of solid dispersions. This article reviews critical aspects and recent advances in
Four generation-to-generation development
Poorly water-soluble drug
formulation, preparation and characterization of solid dispersions as well as in-depth pharmaceutical
Problems and issues solutions to overcome some problems and issues that limit the development and marketability of solid
Mechanism of drug release dispersion products.
Preparation and characterization Ó 2013 Elsevier B.V. All rights reserved.
Future perspectives and strategies

1. Introduction development such as salt formation [7], prodrug formation [8],


particle size reduction [9], complexation [10], micelles [11], micro-
The poor aqueous solubility and dissolution rate of API is one of emulsions [12], nanoemulsions [13], nanosuspensions [14], solid–
the biggest challenges in pharmaceutical development and is lipid nanoparticle [15] and solid dispersion which is considered
becoming more common among new drug candidates over the one of the most successful strategies to improve the dissolution
past two decades due to the use of high throughput and combina- profile of poorly soluble drugs. The term solid dispersions has been
torial screening tools during the drug discovery and selection defined as a dispersion of one or more API in an inert carrier or ma-
phase [1–3]. According to the Biopharmaceutics Classification trix at the solid state prepared by solvent, melting or solvent–melt-
System (BCS), a drug compound is poorly soluble if the highest ing method [16]. The API in solid dispersions can be dispersed as
dose strength is not soluble in 250 ml aqueous media over the separate molecules, amorphous particles, or crystalline particles
pH ranges at 37 °C [4]. These compounds mostly belong to Class while the carrier can be in the crystalline or amorphous state.
II (IIa or IIb), which are poorly soluble and highly permeable Numerous studies on solid dispersions have been published and
according to the pH of the gastrointestinal fluid and tend to present have showed many advantageous properties of solid dispersions
solubility or dissolution rate-limited absorption [5]. Despite their in improving the solubility and dissolution rate of poorly water-
high permeability, these drugs often have low oral bioavailability soluble drugs. These advantages include reducing particle size,
because of their slow and limited release of drug in gastrointestinal possibly to molecular level, enhancing wettability and porosity,
fluid [6]. Therefore, one of the major challenges of the pharmaceu- as well as changing drug crystalline state, preferably into amor-
tical industry is to apply strategies that improve the dissolution phous state [6].
and/or apparent solubility of poorly soluble drugs to develop such Despite such high active research interests, the number of
problematic compounds into orally bioavailable and therapeutic marketed products arising from solid dispersion approaches is dis-
effective drugs [3,5]. appointingly low. This low number is mainly due to scale-up prob-
Various approaches to overcome the poor aqueous solubility of lems and physicochemical instability in the manufacturing process
drug candidates have been investigated in drug research and or during storage leading to phase separation and crystallization
[17–20]. Only a few commercial products have been marketed dur-
ing the past half-century (Table 1). Therefore, in-depth knowledge
⇑ Corresponding author. Bioavailability Control Laboratory, College of Pharmacy, that has been acquired on various aspects of solid dispersions such
Ajou University, Suwon 443-749, Republic of Korea. Tel.: +82 31 219 3442; fax: +82 as carrier properties, preparation methods, physicochemical char-
31 212 3653.
acterization techniques as well as the pharmaceutical mechanism
E-mail address: beomjinlee@gmail.com (B.-J. Lee).

0939-6411/$ - see front matter Ó 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.ejpb.2013.09.007
800 C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813

Table 1 solid dispersions, a crystalline drug is dispersed within a crystal-


Marketed products using solid dispersions [5,72,118]. line carrier forming a eutectic or monotectic mixture [16,21]. In
Product Model drug Carrier type Dosage form the eutectic mixture, the melting point of the mixture is lower than
Certican Ò
Everolimus HPMC Tablet the melting point of the drug and carrier whereas in the monotec-
CesametÒ Nabilone PVP Tablet tic mixture, the melting point of the carrier and drug are constant.
Gris-PEGÒ Griseofulvin PEG Tablet The eutectic mixture is always more preferable because both the
IsoptinÒ SR-E Verapamil HPC/HPMC Tablet drug and carrier will crystallize simultaneously in cooling process,
NivadilÒ Nivaldipine HPMC Tablet
RezulinÒ Troglitazone HPMC Tablet
resulting in a well-dispersed state of the drug in carrier, thus
KaletraÒ Lopinavir, Ritonavir PVPVA Tablet enhancing the dissolution rate. Moreover, the process temperature
IntelenceÒ Etravirine HPMC Tablet for melting eutectic mixture is lower than that of monotectic
ZelborafÒ Vemurafenib HPMCAS Tablet mixture. However, if the mixture of the drug and carrier is not ex-
IncivekÒ Telaprevir HPMCAS-M Tablet
actly at the eutectic composition, the solid dispersion will contain a
CrestorÒ Rosuvastatin HPMC Tablet
AfeditabÒ CR Nifedipin Poloxamer/PVP Tablet mixture of the microfine dispersion and another component as a
FenoglideÒ Fenofibrate PEG Tablet separate phase because one component will progressively crystal-
PrografÒ Tacrolimus HPMC Capsule line until the eutectic composition is reached. In fact the number of
SporanoxÒ Itraconazole HPMC Capsule studied solid dispersions having the exact eutectic composition is
very limited [22]. The API in crystalline solid dispersions may also
exist in amorphous particles or separate molecules (crystalline so-
of matrix formation and drug release are very important to ensure lid solutions). In crystalline solid solutions, the drug molecules can
the preparation of a productive and marketable solid dispersion. replace carrier molecule in the crystal lattice (substitutional crys-
The aim of this review is to provide new knowledge from recent talline solid solutions) or occupy the interstitial spaces between
advances on solid dispersion areas to overcome some problems the solvent molecules in the crystal lattice (interstitial crystalline
and issues that limit the marketability of solid dispersion products. solid solutions) [23].
As a continued work of previous reviews in this field, this article Crystalline carriers in the first generation solid dispersions com-
newly suggests the four classifications of solid dispersions prise of urea [21] and sugars such as sorbitol and mannitol [24].
according to the development by generation that has been investi- These carriers, especially sugars, have high melting point which
gated so far. Finally, the future perspectives and strategies of solid is not favorable for preparing solid dispersions by melting method.
dispersions are also discussed. Urea exhibits high solubility in water and many common organic
solvents while sugars have poor solubility in most of organic
2. The classification of solid dispersions solvents; therefore, sugars were less commonly used than other
carriers. The particle size reduction, wettability improvement and
Depending on the physical state of the carrier which is crystal- polymorphic change are the main reasons for enhancing drug
line or amorphous, the solid dispersions are divided into crystalline solubility and dissolution rate. Zajc et al. [25] prepared the solid
solid dispersions and amorphous solid dispersions respectively. dispersions of nifedipine and mannitol with different ratios by
The solid dispersions can also be classified into four generations melting method. The results showed markedly enhanced dissolu-
based on their composition (Fig. 1). tion rate of nifedipine in comparison with physical mixture
although the crystalline state of nifedipine did not change. The
2.1. First generation SEM results elucidated the mechanism of dissolution rate enhance-
ment which is improving the wettability of nifedipine crystals.
The first generation solid dispersions are crystalline solid Okonogi et al. [26] also tried to improve the dissolution rate of
dispersions. Sekiguchi and Obi [21] prepared the first solid disper- ofloxacin by solid dispersion systems with urea and mannitol.
sions for pharmaceutical application in which urea was used as a The dissolution rate of ofloxacin was significantly increased in urea
carrier to form eutectic mixture with sulphathiazole. In crystalline and mannitol based solid dispersions. Urea based solid dispersions

Poorly water soluble drug

+CC +AP +SFP + WIP or SP

+WIP

+SF +WIP
1stgeneration 2nd generation 3rd generation 4th generation

+SF

- Low dissolution - Dissolution rate - Highest dissolution - Dissolution rate


rate due to crystalline - Precipitation under rate - Controlled release
carrier supersaturation - Precipitation
- Low stability - Low stability under
supersaturation
- Stability

Fig. 1. Composition and properties of four generations of solid dispersions. CC: crystalline carrier, AP: amorphous polymer, SFP: surfactant polymer, WIP: water insoluble
polymer, SP: swellable polymer, SF: surfactant, ("): increase, (;): decrease.
C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813 801

includes povidone (PVP) [33], polyethylene glycol (PEG) [34],


crospovidone (PVP-CL) [35], polyvinypyrrolidone-co-vinylacetate
(PVPVA) [34], and polymethacrylates [36]. The natural product
based polymer consists of cellulose derivatives such as hydroxy-
propylmethyl cellulose (HPMC) [37], hydroxypropylcellulose
(HPC) [38], hydroxypropylmethylcellulose phthalate (HPMCP)
[39], hydroxylpropylmethylcellulose acetate succinate (HPMCAS)
[40], starch (corn starch, potato starch) [41] and sugar glass (treha-
lose, sucrose, inulin) [42]. Among them, HPMC, PEG and PVP are
most widely used [41]. Unlike other amorphous polymers listed
above, PEG has semi-crystalline structure but very low melting
point (below 65 °C) regardless of its molecular weight. Therefore,
PEG is very suitable for the preparation of solid dispersions by
the melting method especially when the drug is heat sensitive. In
contrast, PVP has relatively high glass transition temperature so
it is hardly utilized for the melting method [23]. All of these poly-
mers are soluble in some organic solvents or even wide variety of
solvents in the cases of PEG, PVP, HPC; therefore, these polymers
can be applied to prepare solid dispersions by the solvent method.
Except crospovidone, all of these polymers have high solublility in
water and thus can improve the drug’s wettability when the solid
Fig. 2. Modeling and comparison of dissolution profiles of four generations of solid
dispersions in supersaturated conditions, (0) pure API, (1) the first generation solid dispersions are dispersed in water. Although crospovidone is not
dispersions which show improvement in dissolution rate compared to the pure API, soluble in water, this hydrophyllic polymer quickly swells when
(2) the second generation solid dispersions which have improved dissolution profile contact to water and enhances the drug release rate [35]. The aque-
compared to the first generation thanks to their faster dissolution rate, (3) the third ous solubility of these polymers often decreases and the viscosity
generation solid dispersions which have improved dissolution profile compared to
becomes higher when their chain lengths or molecular weights in-
second generation thanks to their faster dissolution rate and lower precipitation
rate and extend in supersaturated state, (4) the fourth generation solid dispersions crease. The high viscosity polymers can prevent the recrystalliza-
which have improved dissolution profile in a controlled or zero-order manner. tion of drugs in the preparation, storage and dissolution process.
However, they can hinder the dispersion of drugs in carriers in
the preparation process and delay the dissolution rate of drugs
showed higher solubility and dissolution rate than mannitol based when the solid dispersions are dispersed in water. Therefore, the
solid dispersions because urea reduced the crystallinity of ofloxa- molecular weight of polymers is very important factor to select a
cin more than mannitol which was proved by PXRD and DSC polymer as a carrier. In fact, PEG with molecular weights of
results. The main disadvantage of crystalline solid dispersions is 1500–20,000 and PVP with molecular weights of 2500–50,000
the high thermodynamic stability of the carriers that lowers their (K12 to K30) are commonly used in solid dispersions [23]. PEG
dissolution rate compared to amorphous solid dispersions (Fig. 2). with a very low molecular weight can make the solid dispersions
too soft and sticky to manufacture tablets or capsules whereas
PVP with very high molecular weight will reduce the drug release
2.2. Second generation rate because of its low solubility and high viscosity [43]. The inter-
action between the drug and carrier is also important for carrier
The second generation solid dispersions contain amorphous selection. Some drugs have plasticizing effect on polymer or form
carriers which are mostly polymers. Amorphous solid dispersions hydrogen bonds with carriers. These interactions can affect the
can be classified into amorphous solid solutions (glass solutions) preparation process, the drug dissolution rate, the physical proper-
and amorphous solid suspensions according to the physical state ties and the stability of the solid dispersions. Karavas et al. [44]
of the drug. In amorphous solid solution the drug and amorphous prepared felodipine solid dispersions by using different types of
carrier are completely miscible to form molecularly homogenous PVP and PEG as carriers. FTIR data showed hydrogen bonding be-
mixture while amorphous solid suspension consists of two sepa- tween felodipine and both polymers. In the felodipine–PVP disper-
rate phases. Amorphous solid suspensions are formed when the sions, the drug existed as amorphous nanoparticles whereas in
drug has limited carrier solubility or an extremely high melting felodipine–PEG dispersions, the drug was dispersed as crystalline
point [27]. In these systems, the small drug particles mainly in microparticles. This was explained by the higher hydrogen bonding
amorphous state are dispersed in amorphous carriers. It is impor- in the FELO–PVP complex compared to FELO–PEG complex. In that
tant to note that the API often exists more than one state in solid research, the physical state (amorphous or crystalline) and the par-
dispersions, for instance, an API can both dissolve and suspend in ticle size of felodipine in the solid dispersions were determined by
a carrier or exist in amorphous and crystalline state at the same the interaction between drug and polymer. Kohri et al. [160] inves-
time. In amorphous solid dispersions, the API is dispersed in very tigated the oral bioavailability of albendazole solid dispersion
small size (molecular, amorphous particle or small crystals) and granule using HPMC, HPMCP and mixtures thereof as carriers.
exists in supersaturated state in amorphous carriers because of The results showed that all formulations significantly improved
forced solubilization [28–30]. The amorphous carriers can also the dissolution profile of albendazole and the solid dispersions
increase the wettability and dispersibility of drugs as well as containing both HPMC and HPMCP as carriers not only had excel-
inhibit the precipitation process of drugs when amorphous solid lent solubility but also prevented albendazole crystallization in a
dispersions dissolved in water [31,32]. These properties along with neutral medium for 8 h. The absorption study of orally adminis-
the fast dissolution rate of amorphous carriers due to the low ther- tered albendazole in rabbits with low gastric acidity showed more
modynamic stability of amorphous state carriers enhance the drug than three times higher bioavailability of HPMC–HPMCP based so-
solubility and release rate. lid dispersions compared to physical mixtures. In fact, HPMC and
Amorphous carriers can be divided into full synthesis polymers its derivatives have been successfully applied in many marketed
and natural products based polymers. The full synthesis polymer solid dispersion products (Table 1).
802 C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813

Some enteric polymers such as HPMCP, HPMCAS, EudragitÒ L in amorphous and crystalline state. All solid dispersions signifi-
can be used to avoid drug release in the stomach. Since the major- cantly improved the dissolution of ibuprofen and ketoprofen. Inu-
ity of drug absorption occurs in the small intestine and the fast re- tec SP1 is a derivative of inulin prepared by the reaction between
lease of drugs in the stomach can cause precipitation due to drug isocyanates and the polyfructose backbone and can be used as an
supersaturation, the gastric juice resistant polymers may be help- emulsifier in pharmaceutical formulations [52,53]. It was intro-
ful to improve the in vivo bioavailability of drug. Zhang et al. [40] duced into solid dispersions as a novel carrier in the research of
prepared amorphous fenofibrate (FB) solid dispersions using solu- Van den Mooter et al. [20] and showed potential in improving drug
plusÒ, HPMCE5 as carriers for immediate release and HPMCP55, release.
HPMCAS as carriers for enteric release. The in vitro dissolution test Other surfactants and emulsifiers such as sodium lauryl sulfate
showed more improvement in dissolution profile of FB-HPMCE5 (SLS) [54], Tween 80 [55,56], d-alpha tocopheryl polyethylene
and FB-HPMCAS solid dispersions. Although the maximum concen- glycol 1000 succinate (TPGS 1000) [57], polyoxyethylene hydroge-
tration of the drug in the supersaturation dissolution profile of FB- nated castor oil [58] and sucrose laurate [59] are used as additives
HPMCE5 and FB-HPMCAS solid dispersions were similar, the in vivo in solid dispersions. De Waard et al. [60] incorporated SLS in the
pharmacokinetic study in rats showed significant improvement in sugar glass based solid dispersions and prepared tablets. Physical
bioavailability of FB-HPMCAS granules in comparison with FB- mixture of SLS and sugar glass based solid dispersions was also
HPMCE5 granules. This was explained by the complete dissolution compressed to make standard tablets. The dissolution of tablets
of FB in the proximal small intestine and the stronger effect in prepared from solid dispersions in which SLS was incorporated
remaining drug supersaturation state of the FB-HPMCAS solid was strongly improved compared to standard tablets. It was ex-
dispersion. plained that SLS interacted with drug and carrier when it was
Sugar glasses such as trehalose, sucrose and inulin are also used incorporated in solid dispersions; therefore, the dissolution of
to prepare solid dispersions thanks to their extremely fast dissolu- SLS was prolonged and this helped to maintain the high concentra-
tion rate. However, the rapid dissolution of carriers can result in a tion of the drug in the near vicinity of the dissolving tablet during
very high concentration of drugs in the near vicinity of the dissolv- the whole dissolution process, thus preventing the drug precipita-
ing solid dispersions which leads to precipitation and formation of tion effectively.
large drug crystals [42]. Inulin dissolves more slowly than sucrose
and trehalose because of its oligomeric nature; therefore, the pre- 2.4. Fourth generation
cipitation and crystallization rate of inulin based solid dispersions
dispersed in water is lower than trehalose and sucrose based solid The fourth generation solid dispersion is a controlled release so-
dispersions [45]. lid dispersion (CRSD) containing poorly water-soluble drugs with a
short biological half-life. CRSD of poorly water-soluble drugs often
2.3. Third generation requires two targets: solubility enhancement and extended release
in a controlled manner. In CRSD, the molecular dispersion of poorly
Although the amorphous solid dispersions can enhance drug water-soluble drugs in carriers will improve the drug solubility
release rate, the subsequent supersaturation state of drugs may while water insoluble polymers or swellable polymers can be used
cause drug precipitation and decrease the drug concentration to retard the drug release in the dissolution medium [61]. The
in vitro or in vivo, thus negatively affecting the bioavailability of CRSD can deliver an adequate amount of drug for an extended per-
drugs. This phenomenon is very common, especially with sugar iod of time and thus offer many advantages such as improved pa-
glass based solid dispersions. The drug may also recrystallize from tient compliance due to reduced dosing frequency, decreased side
amorphous state in the preparation process (cooling or solvent re- effects, more constant or prolonged therapeutic effect for poorly
moval) and during storage. In the third generation solid disper- water-soluble drugs [62,63]. The conventional polymers used for
sions, the surface active agents or self-emulsifiers are introduced retarding the release of poorly water-soluble drugs in CRSD include
as carriers or additives and showed significant improvement in ethyl cellulose (EC) [62], HPC [28], EudragitÒ RS, RL [61,64],
overcoming the above problems such as precipitation and recrys- poly(ethylene oxide) (PEO) and carboxyvinylpolymer (CarbopolÒ)
tallization. These surfactants are used as process aids and additives [65]. These polymers which are insoluble or dissolve very slowly
which act to improve the biopharmaceutical performance of the in water can sustain the release of poorly water-soluble drugs.
supersaturation systems. The introduction of surfactants or emul- The CRSD uses these sustainable polymers or combines these poly-
sifiers in solid dispersions improves not only the dissolution profile mers in solid dispersion systems or can use other materials having
(Fig. 2) but also the physical and chemical stability of drug. In fact, solubilizing capacity and sustaining action. The CRSD have two
surfactants with amphiphilic structure can enhance the miscibility main mechanisms by which the drug can be released: diffusion
of drugs and carriers and thus reduce the recrystallization rate of and erosion. Cui et al. [64] prepared sustained-release nitrendipine
drugs. Moreover, surfactants or emulsifiers are able to improve microspheres having solid dispersion structure. HPMCP-55 and
the wettability of drugs and prevent the drug precipitation due Aerosil were used as the solid dispersing agents to improve the dis-
to supersaturation by absorbing to the outer layer of drug particles solution of nitrendipine while EudragitÒ RS PO and EC were se-
or forming micelles to encapsulate drugs. lected as the retarding agent to control the drug release rate. The
The surfactants used as carriers include poloxamer [46], compr- bioavailability results in dogs showed the superior prolonged
itol 888 ATO [47], gelucire 44/14 [48], inutec SP1 [20] and absorption profile and bioavailability improvement of solid disper-
soluplusÒ [49]. Poloxamer is a very common polymer which has sion microspheres compared to the reference tablets (Baypress™)
been used for such a long time in solid dispersions as a carrier or and the conventional tablets. Ohara et al. [66] investigated the dis-
additive while soluplusÒ has just been applied in recent years solution mechanism of indomethacin from extended release solid
and showed superior performance in hot melt extrusion processes dispersion system with EC and HPMC (1:1). It was concluded that
[50]. Ali et al. [51] prepared ibuprofen and ketoprofen solid disper- the hydrophobic interaction between indomethacin and EC oc-
sions at different mole ratios using poloxamer 407 and 188. FTIR curred under lower pH region and strongly delayed the dissolution
indicated the disruption of the ibuprofen, ketoprofen dimer con- of indomethacin. Tran et al. [67] prepared polyethylene oxide
comitant with hydrogen bond formation between the drug and (PEO)-based CRSD containing aceclofenac, GelucireÒ 44/14, polox-
carrier. Solid dispersions with mole ratio of drug–carrier 2:1 had amer 407 and pH modifier (Na2CO3) for controlled release of poorly
no drug crystal while at higher drug loading the drug co-existed water-soluble drug. The CRSD improved the dissolution profile of
C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813 803

aceclofenac with an aid of surfactant and pH modifier compared to egies. Solid dispersions can reduce the drug particle size into
pure drug and showed retard drug release in a zero-order manner molecular levels while other conventional particle size reduction
due to the water swellable property of PEO. techniques have particle size limit around 2–5 lm, which easily
agglomerate in the formulation, dissolution process or during stor-
age [69–71]. Novel nanosizing approaches can produce drug nano-
3. Mechanism of drug release from solid dispersions
crystals in a solid state but the preparation process is quite long,
complicated and requires stabilizers as well as special equipments
There are two main mechanisms of drug release from immedi-
[72]. In solid dispersions, the interaction between the drug and car-
ate release solid dispersions: drug-controlled release and carrier-
rier prevents the agglomeration of drug particles and the drug is
controlled release. When solid dispersions are dispersed in water,
released in supersaturation state which is advantageous for rapid
the carriers often dissolve or absorb water rapidly due to their
absorption. Even when the drug from solid dispersions precipitates
hydrophilic property and form concentrated carrier layer or gel
due to supersaturation in the dissolution media, the size of precip-
layer in some cases. If the drug dissolves in this layer and the vis-
itated particles are still in submicron (<1 lm) and the dissolution
cosity of this layer is high enough to prevent the diffusion of the
rate of these particles is still higher than microparticles prepared
drug through it, the rate limiting step will be the diffusion of the
by other techniques [23,73]. However, the precipitated drugs
carrier into the bulk phase and this mechanism is carrier-con-
may also exist in amorphous or different crystalline forms which
trolled release. If the drug is insoluble or sparingly soluble in the
have lower thermodynamic stability (higher energy state) than
concentrated layer, it can be released intact to contact with water
drug particles, leading to faster dissolution for in vivo situations.
and the dissolution profile will depend on the properties of drug
Moreover, solid dispersions can be formulated in solid oral dos-
particles (polymorphic state, particle size, drug solubility) [22]. In
age forms which are more acceptable for patients than liquid prod-
fact, these two mechanisms often occur simultaneously because
ucts produced by solubilization method [70,73]. The preparation
the drug may be partly soluble or entrapped in the concentrated
process of solid dispersions is also more simple and applicable than
carrier layer. However, these mechanisms help explain the differ-
some other techniques such as salt formation or nanosized prepa-
ent release behaviors of solid dispersions and figure out the way
ration (nanoparticle, nano-emulsions).
to improve the dissolution profile of solid dispersions. Numerous
Solid dispersions also have other advantages in improving the
researches showed the improvement of drug dissolution profile
drug bioavailabilty such as wettability, porosity enhancement
when the ratio of carriers in solid dispersions was increased be-
and polymorphic change. The fast dissolution or water absorption
cause the drug was dispersed better and the drug crystallinity de-
of carrier molecules surrounding drug particles can improve the
creased [26]. In these solid dispersions the main release
drug wettability, especially when surfactants or emulsifiers are
mechanism is drug-controlled release. In contrast, other researches
incorporated in solid dispersions. This reduces the risk of agglom-
demonstrated the decrease in drug dissolution rate when the ratio
eration of drug particles in the dissolution media and increase the
of carrier in solid dispersions was increased [68]. This can be ex-
dissolution rate of drug. Many polymers can act as solubilizers to
plained by the carrier-controlled mechanism in which the gel or
enhance drug solubility. Solid dispersions also have high porous
concentrated carrier layer is formed and acts as a diffusion barrier
structure, especially those prepared by solvent method. The fast
to delay drug release. The release mechanism may also be affected
solvent removal may leave some channels in the solid dispersion
by the ratio of drug–carrier in solid dispersions. Karavas et al. [44]
structure, thus increasing the porosity, surface specific area and
prepared felodipine solid dispersions by using different types of
the dissolution rate of drug. Linear polymer based solid dispersions
PVP, PEG as carriers and concluded that the proportion of the drug
often have higher porosity than reticular polymer based solid dis-
in solid dispersions determined the mechanism of drug release
persions [74]. Zhang et al. [40] when preparing amorphous FB solid
which was drug diffusion (through the polymer layer)-controlled
dispersions by thin film freezing techniques as mentioned above
at low drug contents and drug dissolution-controlled at high drug
also found that the thin film freezing process dramatically in-
contents. Therefore, in order to improve the dissolution profile of
creased the surface area of the aggregates of FB-polymer composi-
solid dispersions, it is important to identify the mechanism release
tions by at least 40-times compared to the bulk FB powder and the
of solid dispersions rather than only focus on the polymorphic
significant increase in surface area facilitated the enhancement of
state of drugs because in carrier-controlled release solid disper-
dissolution rate and bioavailability of FB. Numerous researches
sions, the carrier properties such as solubility, viscosity, gel form-
on solid dispersions demonstrated the change in polymorphic state
ing ability and the ratio of drug–carrier are the key factors
of drugs from crystalline to amorphous form. In general, the reduc-
affecting the drug dissolution profile.
tion of drug crystallinity will improve the dissolution rate because
In CRSD, depending on the characteristic of polymers and the
less energy is required to break up the crystal lattice during the
miscibility of the drug and carrier there are two main mechanisms
dissolution process, thus increasing the drug solubility [33]. The
by which the drug can be released from the system: diffusion and
polymorphic state of drugs in solid dispersions is mainly
erosion. If the drugs and polymers are well dispersed in internal
determined by the preparation process and the physicochemical
structure of solid dispersions, the diffusion of drugs through the
interactions between drug and carrier.
matrix will be the main mechanism. If the drugs and carriers exist
in separated particles, the solid dispersion erosion may become the
main mechanism for drug release. In some solid dispersions, both
5. Common problems of solid dispersions
of these mechanisms can control the drug release at the same time
[66].
Despite many advantages in improving dissolution profile of
poorly water soluble drugs, the number of commercial products
4. Advantages of solid dispersions using solid dispersions is limited because of some problems in
the preparation process and during storage. The most important
In comparison with other techniques used to improve bioavail- problem is the recrystallization of drugs from amorphous state
ability of poorly water soluble drugs such as salt formation, during storage which leads to decreased bioavailability of solid
particle size reduction (milling or micronization), and solubiliza- dispersions. The crystallization of drugs involves two steps: nucle-
tion (cosolvent, micelles, emulsions), solid dispersions show many ation followed by crystal growth that requires the diffusion or rear-
important advantages to become one of the most promising strat- rangement of drug molecules [1]. Therefore, molecular mobility of
804 C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813

drugs is the key factor determining the drug physical stability. The bonding between the drug and polymer proved by FTIR data. No
molecular mobility can be divided into global mobility (a-relax- hydrogen bond between the griseofulvin and PVP was noticed;
ations) which is the molecular rotation, translation for the glass therefore, the author concluded that phase separation rates ob-
transition and local mobility (b-relaxations) which relates to the tained were proportional to the drug–polymer interactions and
movement of only a part of a molecule and occurs at a smaller time the drug content of the dispersion. In many researches, although
scale than global mobility [3,75]. At 50 °C below the glass drugs existed in crystalline state in solid dispersions, the in vivo
transition temperature (Tg), the a-relaxations are negligible and bioavailability was increased significantly in comparison with
b-relaxations are thought to determine the rate of nucleation and physical mixture. These solid dispersions will not face crystalliza-
crystallization of amorphous materials [76–78]. The presence of tion problems and thus have good stability [51,84].
water often reduces the Tg of amorphous systems due to its plas- The appearance of surfactants and emulsifiers in third genera-
ticizing effect and thus increases the molecular mobility [79,80]. tion solid dispersions may increase the miscibility of drug–poly-
The increased temperature also enhances the molecular mobility mer due to their amphiphilic structure and thus reduce the
and causes recrystallization in solid dispersions. Therefore, the recrystallization rate. The surface active agents can also reduce
storage temperature and moisture are important factors affecting the process temperature of melting method due to their plasticiz-
the physical stability of drug. The poor scalability for the purposes ing effect and increase the solubility of drugs and carriers in organ-
of manufacturing is also the main obstacle for solid dispersions to ic solvents in the solvent method. When solid dispersions are
be available in the market [6]. dispersed in water, the surface active agents improve drug wetta-
Furthermore, there are some common problems in the prepara- bility and inhibit the precipitation process by absorbing on the sur-
tion, formulation and dissolution process that researchers have to face of drug particles or forming micelles encapsulating drugs.
consider in research and development of solid dispersion products. Mura et al. [85] investigated the effect of anionic surfactants
These problems include the thermal instability of drugs and including SLS, dioctylsulfosuccinate (DSS) and nonionic surfactant
carriers in the melting method, the solvent residue in the solvent (Tween 60) on the properties of ketoprofen ternary solid disper-
method, the recrystallization of drugs in solidification and further sions in polyethylene glycol 15,000. Although the binary system
formulation process, the low in vivo–in vitro correlation, and the (ketoprofen–PEG) showed the dissolution enhancement compared
precipitation of drugs after dissolving in water due to supersatura- to pure drug, the dissolution profile of solid dispersion was signif-
tion. Ayenew et al. [81] investigated the effect of compression in icantly improved when the surfactants were incorporated to make
the tableting process on miscibility of naproxen–PVP K25 solid dis- ternary systems. It was explained by the presence of the surfac-
persions. FTIR showed that the miscibility in the compositions with tants which improved wettability and decreased surface tension
30% and 40% (w/w) naproxen is markedly affected by the compres- of drug particles and dissolution medium, leading to the inhibition
sion process. Compression pressures from beyond 565.05 MPa in- of drug precipitation. The micellar solubilization effect of surfac-
duced apparent amorphous–amorphous phase separation. This tant was excluded because the surfactant concentration in the dis-
was explained by the weakening and/or disruption of intermolec- solution medium was generally below the critical micelle
ular hydrogen bonding between drug and polymer upon compres- concentration. The crystallinity degree of the ternary systems
sion. It is important to mention the poor correlation between was also lower than binary system which showed the efficiency
in vitro dissolution data and in vivo absorption despite the use of of surfactants in improving drug–polymer miscibility. The effec-
biorelevant in vitro dissolution media in case of poorly water-solu- tiveness of the incorporated surfactants depended on the types of
ble drugs [82]. Although SUPAC-based biowaiver is possible by FDA surfactants (SDS > DSS > Tween 60) and the solubilizing efficiency
in case of BCS Class I, in vivo testing or a clinical assessment would of anionic surfactants was higher than the nonionic one. The great-
be always needed for a BCS Class II (even III, IV) compound to prove est efficacy of SDS could be due to its high hydrophilic property
the efficiency of solid dispersions in improving bioavailability of and strong solubilizing ability.
poorly water soluble drugs. In order to prevent the recrystallization of drugs in the solidifi-
cation process (cooling, solvent removal) many techniques have
been developed to increase the solidification rate such as ultra ra-
6. Strategies to overcome the common problems of solid pid freezing, spray drying and spray freezing which will be men-
dispersions tioned below.

Polymers often have higher glass transition temperature (Tg)


7. Preparation method
compared to the API so they can decrease the molecular mobility
of drug by increasing the Tg of the miscible mixture or by interact-
There are three major preparation methods for solid dispersions
ing with drug molecules [33,76]. A polymer has to be miscible with
including melting method, solvent method and melting solvent
drug to prevent the drug recrystallization and the miscibility de-
method (Fig. 3). In fact, the melting method and solvent method
pends on the molecular interaction between the drug and polymer
are more common than the melting solvent method.
[17,83]. Hydrogen bonding between the drug and polymer is the
main force to increase the solid miscibility and thus hinder phase
separation and drug recrystallization. However, many polymers 7.1. Melting method
are hygroscopic and the water absorbed by polymers may increase
molecular mobility. Therefore, selecting a suitable polymer which 7.1.1. Characteristic features, advantages and limitations of melting
has high Tg, strong interaction with drug and low hygroscopicity method
is the way to stabilize the physical state of drugs in solid disper- The first solid dispersions for pharmaceutical application were
sions. Vasanthavada et al. [83] investigated factors influencing so- prepared by melting method or fusion method [21]. In the melting
lid solubility and phase separation kinetics of drugs in method, the drug and carrier are melted together at a temperature
griseofulvin–PVP and indoprofen–PVP solid dispersions. Griseoful- above the eutectic point, which is the lowest possible melting
vin did not exhibit any solid solubility in PVP, whereas a 13% w/w point of the mixture, then the liquid is cooled or solidified by dif-
indoprofen remained molecularly miscible with the polymer under ferent techniques such as ice bath agitation [16], spreading on
accelerated stability conditions of 40 °C/69% RH for 3 months. The stainless steel thin layer cooled by air or water [86], putting petri
higher miscibility of indoprofen in PVP was explained by hydrogen dishes inside a dessicator [87] and immersion in liquid nitrogen
C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813 805

Preparation method
sion. This phenomenon can be prevented in some cases by using
surfactants. The phase separation can also occur during cooling,
as the drug–carrier miscibility changes. Indeed, slow cooling the
mixture often produces drugs in crystalline state, while fast cooling
may yield drugs in amorphous state. This limitation can be con-
Melting Solvent evaporation Melting solvent trolled by using spray congealing technique to quickly solidify
melted mixture in cool air. Spray congealing is a process in which
a hot molten mixture (solution or suspension of a drug in a molten
carrier) is atomized into an environment maintained at tempera-
• Ice bath agitation • Oven drying tures below the carrier melting point [90,91]. The atomization
• Thin film cooling • Vacuum drying makes the melted droplets congeal rapidly to form solid particles.
• Liquid nitrogen • Rotary evaporation This method was used in many researches to prepare solid disper-
• Heating on hot plate
• Spray congealing sions [92,93].
• Spray drying
• Hot-melt extrusion Another limitation of the melting method is the thermostability
• Freeze drying
• MeltrexTM
• Supercritical anti-solvent of the API and the carrier at high temperature. A modified tech-
• Melt agglomeration • Co-precipitation nique to reduce the drug heating time and the process temperature
• Electrostatic spinning was performed by suspending the API in a previously molten car-
• Spray freeze drying rier, instead of heating both drug and carrier at the same time to
• Ultra-rapid freezing
obtain molten mixture. Mehanna et al. [94] applied this technique
• Fluid-bed coating
to prepare tadalafil-PluronicsÒ solid dispersions. PluronicsÒ of var-
ious grades was melted at 70 ± 2 °C, followed by the addition of
drug powder to the molten carrier and stirring until a homogenous
Fig. 3. A branched tree used for the production of solid dispersions. dispersion was formed. The SEM, DSC, PXRD results showed micro-
crystalline uniform dispersion of tadalafil in the carrier matrix.
Although the drug was still in crystalline state, the dissolution rate
[88]. The resultant solid is then crushed, sieved, pulverized to re- of tadalafil in Pluronic F-127 based solid dispersion significantly
duce the particle size or injection molded into dosage forms with- increased compared to the physical mixture. Several important
out undergoing milling. The advantage of this method is that it modifications of melting method to overcome its limitations con-
does not require any solvent. An important prerequisite of this sist of hot-melt extrusion, Meltrex™, and melt agglomeration.
method is the miscibility of the drug and carrier in the molten state
to obtain a homogenous mixture. Therefore, a suitable carrier 7.1.2. Hot melt extrusion
should shares similar physicochemical properties with the API. In recent years, hot melt extrusion has become one of the most
PEG and poloxamer are two polymers that are commonly used to common methods for solid dispersion preparation due to its high
prepare solid dispersions by the melting method. Kolašinac et al. scalability and applicability. In this method, the drug and carrier
[68] prepared solid dispersions by melting method at a low tem- are simultaneously mixed, heated, melted, homogenized and ex-
perature (70 °C) using poloxamer P 188 and poloxamer P 407 to in- truded in a form of tablets, rods, pellets, or milled and blended
crease the solubility and the dissolution rate of desloratadine. Solid with other excipients for different purposes [95]. The intense mix-
dispersions containing different weight ratio of poloxamers and ing and agitation forced by the rotating screw during the process
desloratadine were prepared and then were used for the prepara- cause disaggregation of drug particles in the molten polymer,
tion of immediate release tablet. The results showed that deslorat- resulting in a homogenous dispersion [32]. This process involves
adine existed in the amorpohous state in solid dipersions and both the transformation of a solid mass of intertwined particles into a
types of poloxamer significantly enhanced intrinsic solubility of viscous liquid or semisolid mass by heating and intense mixing.
desloratadine (10–80 times) as well as the dissolution profile of The hot melt extruded systems are composed of drugs, one or more
desloratadine tablets. Poloxamer P 407 was approximately 4-fold meltable polymers and other additives such as plasticizers and pH
less effective than poloxamer P 188 as a solubilizer because of its modifiers. The melting rate is mainly affected by the physical and
higher molecular weight and higher proportion of hydrophobic rheological properties of the polymer. The melting rate is much
polyoxypropylene segment. The increase in poloxamer ratio over faster when polymers are amorphous and low viscous. Hot melt
1:1 in solid dispersions decreased intrinsic dissolution rate of desl- extrusion of miscible components may lead to a high trend of
oratadine because larger amount of poloxamer in solid dispersions amorphous solid dispersion formation, thus improving drug disso-
provide better conditions for gel layer formation when contacting lution profile. In order to select a suitable polymer for hot melt
with water, which consequently acts as a diffusion barrier and de- extrusion process, Hansen solubility parameter can be applied to
lays drug release. The high compression force reduced the dissolu- predict the drug–carrier miscibility [96,97]. Despite some prob-
tion rate of desloratadine by reducing the porosity of the tablet and lems such as the miscibility of drugs and carriers as well as high
increasing bonding between poloxamer particles. Different types of local temperatures in the extruder due to high shear forces, hot
PEG such as PEG 4000 and PEG 6000 were also used as carriers to melt extrusion has considerable advantages for pharmaceutical
increase the solubility of antiviral thiocarboxanilide UC-781 and applications [98]. An important advantage of the hot melt extru-
nimodipine in solid dispersions prepared by the melting method sion method compared to other melting methods is the low resi-
[48,89]. dence time of the drug and carrier at elevated temperature in the
Despite being frequently applied, the melting method has some extruder which reduces the risk of degradation of thermolabile
noticeable limitations that can severely impact attempts to en- drugs [23]. This method is also continuous, efficient, easy scale-
hance the bioavailability of poorly water-soluble drugs. A major up and produce higher thermodynamic stability products than
disadvantage is that the method can only be applied when the drug other methods [99]. This technology has been successfully applied
and the carrier are compatible and miscible at the heating temper- to design many drug delivery systems such as immediate and con-
ature. An incompatibility causes phase separation and the high trolled release tablets, granules, pellets, implants, transdermal
viscosity of a polymeric carrier in the molten stage can prevent drug delivery systems, ophthalmic ocular inserts [100]. Various
the miscibility, thus resulting in an inhomogeneous solid disper- references have applied hot melt extrusion for solid dispersion
806 C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813

preparation. For instance, fenofibrate solid dispersion with carrier carefully considered, because a large excess may induce a signifi-
of Eudragit E100 or polyvinylpyrrolidone-vinyl acetate copolymer cant change in the matrix structure. The disadvantage of this meth-
S630 was prepared by hot melt extrusion technology, which re- od is that the residual solvent remaining after evaporation process
sulted in an increase in dissolution rate and oral bioavailability may cause toxicity and complete solvent removal is nearly impos-
[101]. Hot melt extrusion and its advanced technology, Meltrex™, sible. In addition, residual solvent can also, like water, lower the Tg
are the most successful ones to prepare solid dispersions with and act to plasticize the system, leading to phase separation due to
many marketed products such as CesametÒ, RezulinÒ, KaletraÒ the increased mobility of components. The use of solvent mixtures
(Meltrex™), NovirÒ (Meltrex™) and IsotipÒ (Meltrex™) [102,103]. has been proposed as the way to minimize the problems relating to
organic solvents. For example, Kumar [110] used a dioxane–buta-
7.1.3. Meltrex™ and melt agglomeration nol–water mixture to prepare an amorphous solid dispersion of
Meltrex™ based on the hot melt extrusion principle is a an opioid antagonist. Other disadvantages of the solvent method
patented solid dispersions manufacturing process. In Meltrex™ are its large environmental issues, the high cost of production
technology, the use of a special twin screw extruder and the pres- due to the extra facilities required for solvent removal and protec-
ence of two independent hoppers allow conveying the extruded tion against explosion as well as low scalability. For these reasons,
mass continuously throughout the extrusion channel and thus re- hot melt extrusion is more favorable than solvent method to pre-
duce the residence time of the drug (approximately 2 min) in the pare solid dispersions if the stability and bioavailability enhance-
extruder as well as avoid thermal stress to the drug and excipient ment of solid dispersions prepared by hot melt extrusion are
[6,104]. The temperatures of all the barrels are independent and ensured.
can be accurately controlled from low temperature (30 °C) to high Similar to the melting method, the solidification rate in solvent
temperature (250 °C) [104]. This technique is suitable for drugs method can determine the physical state of drugs in solid disper-
which are sensitive to oxidation and hydrolysis because oxygen sions. A fast solidification method is always preferred to guarantee
and moisture may be eliminated during the process. the amorphous state of drugs. Therefore, there are many methods
In general, the amorphous solid dispersion prepared by melting developed for fast solvent removal such as heating on a hot plate
method are soft, sticky and have poor flow properties and poor [62], vacuum drying [111], rotary evaporation [112], spray drying
compressibility which hinder their applications in a large pharma- [113], freeze drying [38], spray freeze drying [37] and ultra rapid
ceutical scale of tableting [105]. Melt agglomeration method, in freezing [114].
which the carrier acts as a meltable binder, is a feasible method
to solve these problems. Melt agglomeration is processed in high 7.2.1. Vacuum drying and rotary evaporation
shear mixers or rotary processor with the mixture prepared by Phase separation is a challenge that can arise during the solvent
three ways: adding the molten carrier containing the drug to the removal process. The heating process can increase the molecular
heated excipients [106,107], adding the molten carrier to a heated mobility resulting in phase separation. Therefore, vacuum drying
mixture of the drug and excipients [46], or heating a mixture of the and rotary evaporation at a moderate temperature are used to
drug, carrier and excipients to a temperature within or above the avoid that risk and prevent the degradation of drugs and carriers
melting range of the carrier [106]. The rotary processor may be at high temperature. After solvent evaporation process, the resul-
preferable to the high shear mixer in melt agglomeration tech- tant solid dispersions may be stored in a vacuum desiccator for
nique because the temperature can be more easily controlled and complete removal of residual solvent. Although these methods
higher binder content can be incorporated in the agglomerates are easy to perform, the processes are quite long. Therefore, the
[29]. Seo et al. [106] prepared the agglomerates containing solid phase separation and recrystallization of drugs can easily occur
dispersions of diazepam by melt agglomeration in a high shear during the drying process.
mixer. This study showed significant increase in dissolution rate
of diazepam and a higher dissolution rate was obtained with a 7.2.2. Spray drying
lower drug concentration indicating a higher degree of molecular Spray drying is an efficient technology for solid dispersion man-
dispersion at the lower concentration. It was concluded that the ufacturing because it permits extremely rapid solvent evaporation
dissolution rate was affected mainly by the type of meltable binder resulting in fast transformation of an API-carrier solution to solid
since Gelucire 50/13 gave rise to faster dissolution rate compared API-carrier particles [115]. In this technique, the API-carrier solu-
to PEG 3000. tion or suspension is transported from the container to the nozzle
entrance via a pump system and atomized into fine droplets with
7.2. Solvent evaporation method (solvent method) large specific surface area. These droplets result in rapid evapora-
tion of the solvent and the formation of solid dispersions within
In the solvent evaporation method, solid dispersion is obtained seconds. The size of the solid dispersion particles prepared by
after the evaporation of solvent from the solution containing a spray drying can be customized by modulating the droplet size
drug and carrier [16]. The solvent method has solved main prob- via nozzle to meet the requirements for further processing or
lems of the melting method relating to the decomposition of drugs applications. The drugs in solid dispersions prepared by spray dry-
and carriers at high temperature because in the solvent method, ing are often in amorphous state; therefore, the solubility and dis-
the solvent removal can be performed without heat such as freeze solution rate are significantly increased. Spray drying is one of the
drying technique. Some polymers hardly used as carriers in the most common techniques used to prepare solid dispersions due to
melting method due to their high melting point can be applied in the possibility of continuous manufacturing, ease of scalability,
the solvent method. An important prerequisite of this method is good uniformity of molecular dispersion and cost-effectiveness in
the sufficient solubility of the drug and carrier in a solvent or co- large scale production with high recoveries (more than 95%)
solvent [23]. Finding a suitable non-toxic solvent is sometimes [116–118]. Solid dispersion products prepared by spray drying
difficult because carriers are hydrophilic whereas drugs are hydro- are commercially available such as IncivekÒ and IntelenceÒ [118].
phobic [108]. The solvents used in solvent method may include
methanol, ethanol, ethyl acetate, methylene chloride, acetone, 7.2.3. Freeze drying
water. . .and mixtures thereof [109]. Some surfactants such as Freeze drying is an alternative method for drying solid
Tween 80 and SLS can be utilized to increase the solubility of drugs dispersions. This method shows promise as a suitable technique
and carriers in solvents. However, their incorporation has to be for the incorporation of drugs into stabilizing matrices because
C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813 807

the drug is subjected to minimal thermal stress during the solid critical fluids allow the precise control of the solubilization of
dispersion formation. This method includes two steps: freezing many drugs. When using CO2 as a solvent, the drug and carrier
and lyophilization. The freezing rate is very important to control are dissolved in supercritical CO2 and sprayed through a nozzle
phase separation. The basic process consists of immersing the into an expansion vessel with lower pressure. The rapid expansion
API-carrier solution in liquid nitrogen until it is fully frozen and induces rapid nucleation of the dissolved drugs and carriers, lead-
the frozen solution is then lyophilized [27,119]. The advantage of ing to the formation of solid dispersion particles with a desirable
this method is the minimized risk of phase separation and the dis- size distribution in a very short time. When supercritical CO2 are
advantage is that most organic solvents have low freezing temper- used as anti-solvent, it is introduced into the nozzle simulta-
atures and do not stay frozen during sublimation. Furthermore, neously with drug–carrier solution in an organic solvent. When
sample temperature has to be maintained below the Tg of the the solution is sprayed, the solvent is quickly extracted by the
frozen concentrated fraction when glass formation is required. supercritical CO2 leading to the precipitation of solid dispersion
Therefore, the process will take much longer time if low tempera- particles [58].
ture is required for freeze-drying of a sample. In order to obtain an
acceptable duration for the lyophilization process, the solvent 7.2.6. Co-precipitation method
should have a sufficiently high vapor pressure. Co-precipitation is a suitable technique to prepare solid disper-
sions of poorly water-soluble drugs which have low solubility in
7.2.4. Cryogenic processing techniques commonly used organic solvents and high melting points that can-
Solid dispersions can also be obtained by cryogenic processing not be processed by melting and other solvent methods [128]. In
techniques including spray freeze drying (SFD) and ultra-rapid this method, a drug and carrier are completely dissolved in an or-
freezing (URF). These methods increase the freezing rate compared ganic solvent before being added to an anti-solvent which causes
to freeze drying technique. SFD allows for a reduction in the pri- simultaneous precipitation of the drug and carrier. The resulting
mary particle size of drug particles without intense frictional or suspension is then filtered and washed to remove residual sol-
mechanical forces which can cause degradation of the API through vents. The co-precipitated material obtained after filtration and
thermal stress [120]. SFD is a very promising drying technique in drying is referred to microprecipitated bulk powder (MPD) which
which the solution of the drug and polymer is sprayed into/on li- is a solid dispersion of the drug and carrier [129]. The polymers
quid nitrogen or cold dry air and the frozen droplets are subse- used in co-precipitation method often have pH dependent solubil-
quently lyophilized. The large surface area and direct contact ity such as polymethylacrylate, polymethylmethacrylate, HPMCP,
with the cooling agents result in rapid vitrification, thereby reduc- HPMCAS, polyvinyl phthalate and cellulose acetate phthalate while
ing the risk of phase separation to a minimum. It also offers the some solvents such as dimethylacetamide, dimethylformamide
potential to customize the particle size to make it suitable for fur- and N-methyl pyrrolidone are mainly used due to their excellent
ther processing or applications. SFD usually produces drugs in solvency power, particularly for high molecular weight polymers
amorphous state because the rapid freezing rate can prevent [129]. Thanks to the poor solubility of ionic polymers in a specific
molecular arrangement into crystalline domains [121–123]. More- range of pH, the drug and polymer can be simultaneously precipi-
over, in another research, Van Drooge et al. [124] demonstrated tated in water (as anti-solvent) and washed by aqueous solution to
that the drug stability in SFD solid dispersions is significantly bet- remove residual organic solvents. The advantage of this method
ter than that of comparable freeze dried solid dispersions due to compared to other methods is that it does not need elevated tem-
higher cooling rate of SFD resulting in more incorporation of the peratures which can cause degradation of the drug or carrier. The
drug into matrix, especially at high drug loads. Another cryogenic solvents can be less volatile and can be washed by aqueous solu-
process, URF, involves the use of a solid cryogenic substrate with tion to remove organic solvents from the product [130]. Moreover,
thermal conductivity between 10 and 20 W/(m K) [114]. In this co-precipitation method is applicable in large scale and ZelborafÒ
technique, a drug–polymer solution is applied to a cryogenic solid is the marketed solid dispersion product prepared by this technol-
substrate and the frozen particles are then collected and removed ogy [131]. It is important to mention that the solvent and anti-sol-
the solvent by lyophilization [125]. As the supercooling is extre- vent mixture used in the precipitation process may act as
mely fast, the nucleation of API crystals is minimized or completely plasticizers resulting in high molecular mobility and crystallization
prevented, resulting in amorphous morphology after lyophilization [130]. Therefore, the control of precipitation process and the selec-
[126]. As compared to traditional methods, cryogenic processing tion of suitable polymer, solvent and anti-solvent are critical to the
techniques offer more advantages. Spray drying and melting meth- success of final products. Dong et al. [132] used HPMC-AS to pre-
od are not always appropriate for use with thermolabile drugs. pare solid dispersions of a BCS Class II compound by co-precipita-
However, SFD and URF were performed under sub-ambient condi- tion method and hot melt extrusion for comparison. PXRD results
tions and hence they can avoid heat or air that induces degradation indicated the amorphous state of the drug in both products.
of drugs during drying processing. In a recent research described Although two products have similar true densities, the co-precipi-
by Badens et al. [127], SFD and supercritical anti-solvent (SAS) tation product is more porous and has a larger specific surface area
were compared in preparation of solid dispersions containing than the HME product which may explain for the faster dissolution
oxeglitazar. SFD showed lower crystallinity and higher dissolution profile of co-precipitation product.
rate than SAS. However, SAS is more favorable in terms of time
consumption. 7.2.7. Electrostatic spinning
Electrostatic spinning (electrospinning) can be considered a
7.2.5. Supercritical anti-solvent (SAS) combination of solid dispersion technology and nanotechnology.
SAS method generally uses supercritical carbon dioxide (CO2) as In this method, a drug–polymer solution is placed into a spinneret
a solubilizing solvent or anti-solvent. When supercritical carbon connected with a microsyringe pump and a high voltage between 5
dioxide is utilized as a solvent, this method is considered an envi- and 30 kV is applied to the needle tip to induce a charge on the sur-
ronmentally friendly technique because no organic solvent is re- face of the solution [133]. A fixed electrical potential is also applied
quired. Supercritical fluids are fluids whose temperature and across a fixed distance between the spinneret and the collector
pressure are above the critical point. The favorable properties of [134]. When electrical forces overcome the surface tension of the
gases such as high diffusivity, low surface tension and low viscosity feeding solution at the air interface, polymer jets are ejected
imparted to liquids through manipulation of the pressure of super- [135]. After coming out, the charged jets go straight for some
808 C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813

distance, and then travel a spiral path because of the whipping of crystallinity are importantly characterized. The amount of drugs
instability [133]. As the jet accelerates through the electric field, existing in amorphous state can be calculated indirectly from the
the solvent evaporates rapidly to make fibers at micron or submi- extent of crystallinity in the sample. The crystalline state of drugs
cron diameter which are collected on the screen or a spinning is commonly characterized by the following techniques: thermo-
mandril. The collected fibers produce a non-woven fabric, which analytical techniques such as Differential Scanning Calorimetry
can be used in oral dosage forms by direct incorporation of the (DSC) and Modulated Differential Scanning Calorimetry (MDSC);
materials into a capsule or by further processing such as milling powder X-ray diffraction (PXRD); Confocal Raman Spectroscopy.
or grinding [135]. In this process, the diameter and morphology Other instrumental techniques such as Fourier Transformed Infra-
of the filaments are affected by solution surface tension, the poly- red spectroscopy (FTIR), solid state nuclear magnetic resonance,
mer solution dielectric constant, feeding rate, the electric field Thermal Gravimetry Analysis (TGA) are used to investigate the
strength, tip-to-collector distance as well as some environmental chemical stability and molecular interaction of the drug and car-
parameters such as temperature, humidity and air velocity in the rier. Microscopy techniques such as optical microscopy, transmis-
spinning chamber [134]. The main advantage of this technique re- sion electron microscopy (TEM), scanning electron microscopy
lates to the extremely high surface area per unit mass of fibers (SEM) and atomic force microscopy (AFM) are also used to qualita-
which facilitates the fast and efficient solvent evaporation leading tively characterize the crystalline states of drug, the molecular
to the formation of amorphous dispersions [136]. Therefore, the miscibility, phase separation and surface morphology of solid
dissolution of the API incorporated in these fibers is improved sig- dispersions.
nificantly by two mechanisms: nanosizing and amorphization
[137]. Yu et al. [138] prepared PVP based solid dispersions of acet- 8.1. Differential Scanning Calorimetry (DSC) and Modulated
aminophen using electrostatic spinning and compared with other Differential Scanning Calorimetry (MDSC)
methods. The results showed that electrospun nanofiber-based so-
lid dispersions had significantly faster dissolution profile compared The basic principle of thermal analytical approaches is the
to other casting-film solid dispersions prepared by vacuum drying, dynamic changes in the solid-state properties of material initiated
freeze drying, and heat-drying. by the heating or cooling process. DSC, the most commonly used
thermal technique for solid dispersion characterization, provides
7.2.8. Fluid-bed coating accurate information about melting point, glass transition temper-
In this method, the drug and carrier are firstly dissolved in a ature as well as the energy changes associated with the phase tran-
solvent. This solution mixture is sprayed through a nozzle onto sitions including crystallization and fusion process. The lack of a
the surface of nonpareil pellet in a fluid-bed coater [139,140]. drug melting peak in the DSC thermogram of a solid dispersion
The solvent is removed by drying airflow and the co-precipitate indicates that the drug exists in an amorphous form. In DSC, the
simultaneously deposited on the surface of nonpareil pellets glass transition endotherm, crystallization exotherm and fusion
[141]. The advantage of this method is that solid dispersion endotherm can also be quantified and used to calculate the degree
granules or pellets can be ready for tableting or encapsulating into of crystallinity [143]. For instance, Urbanetz [30] used DSC to eval-
capsules without further handling. As showed in the researches of uate the remaining amorphous content of a drug in solid disper-
Sun et al. [142], solid dispersions prepared by fluid-bed coating sions during storage conditions. However, the degree of
demonstrated the bioavailability improvement of fenofibrate and crystallinity which is under 2% may not be detected by DSC [23].
silymarin in vivo. DSC parameters, such as endotherm (DHtr, J/g), was also used to
predict the bioavailability of solid dispersion products by Berndl
et al. [144]. It was concluded that the solid dispersions of itraco-
7.3. Melting-solvent method
nazole improved bioavailability as DHtr < 0.35 J/g and the lower
endotherm of solid dispersions obtained when the higher energy
The melting solvent method is the combination of the melting
inputted during melt extrusion process led to bioavailability
method and the solvent method. In this method, the drugs are
improvement.
dissolved in a suitable solvent and mixed with the molten carrier
Modulated DSC is an advanced thermal technique that can de-
followed by solvent removal and solidification to form solid disper-
convolute the different thermal events obtained from DSC. In
sions. The advantage of this method is that the temperature and
MDSC, a sinusoidal wave modulation is superimposed on top of
the mixing time are lower than melting method, thus protecting
the conventional linear temperature ramp allowing the separation
the drug from thermal degradation. In addition, the carrier in the
of total heat flow in DSC into reversing and non-reversing thermal
molten state is more easily dispersed and dissolved in the solvent
transition [1]. Thermal events such as enthalpic relaxation, evapo-
in comparison with solvent method.
ration, crystallization, thermal decomposition and some other
A branched tree listed the preparation techniques for solid dis-
melting events are distributed in the non-reversing heat flow while
persion is showed in Fig. 1. In fact, the optimal method and tech-
the reversing heat flow includes other melting events and glass
nique for solid dispersion preparation was chosen based on the
transition [145]. The application of this MDSC technique in solid
physicochemical properties of drugs and carriers. In general, spray
dispersion characterization has become very popular due to its
drying and hot melt extrusion are most commonly used due to
advantages such as the improvement of both sensitivity and reso-
their high scalability and applicability.
lution, analysis of complex overlapping transitions, direct mea-
surement of the heat capacity and detection of weak glass
8. Characterization of physicochemical properties transitions compared to standard DSC [146]. As described by Gui-
not and Leveiller [146], MDSC method was successfully used to de-
The dissolution enhancement of poorly water-soluble drugs in tect and quantify low levels of amorphous phase in crystalline drug
solid dispersions can be proven by the standard dissolution meth- through measurements of the heat capacity jump associated with
ods. Other properties of solid dispersions such as the physical the amorphous phase glass transition. Ghosh et al. [145] tried to
states of drugs, the drug–carrier interaction and the physical and develop a stable amorphous solid dispersion of a poorly water-sol-
chemical stability of drugs should also be evaluated. Consequently, uble and highly thermal sensitive compound by hot melt
many instrumental and analytical techniques are applied to mea- extrusion. Preliminary MDSC studies showed that propylene glycol
sure these properties. The crystalline state of drugs and the degree was the most suitable plasticizer which could provide single phase
C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813 809

solid dispersion of the API with various polymers and also facilitate process and the biocompatibility of loratadine and PVP in solid dis-
low processing temperatures. The molecular miscibility, recrystal- persions. The results showed that loratadine was stable up to
lization and phase separation of solid dispersions after preparation 203 °C at which it started losing mass. The thermoanalytical curves
and under accelerated stability conditions were characterized by of solid dispersions and their components were not significant dif-
MDSC. In this study, a single Tg of solid dispersions showed ferent, which suggested that chemical interactions accelerating
molecular miscibility and a new Tg detected by MDSC indicated drug or polymer degradation did not occur.
phase separation. The solid dispersion system was physically sta- Physicochemical characterization of solid dispersions is
ble if no re-crystallization peaks were observed. essential to evaluate the pharmaceutical applicability of solid dis-
persions and thoroughly understand the pharmaceutical mecha-
8.2. Powder X-ray diffraction (PXRD) nisms of drug dissolution enhancement and physicochemical
stability. Therefore, an ongoing development of new and advanced
PXRD is the most widely used method to identify and character- characterization techniques in solid dispersion area is very
ize the crystalline state of drugs in solid dispersions. This method necessary.
can detect material with long-range order as well as expose sharp
diffraction peaks that indicate crystalline compound with
characteristic fingerprint region. Thanks to the specificity of the 9. Future perspectives and strategies
fingerprint, the drug crystallinity can be separately identified from
the carrier crystallinity and thus can differentiate the amorphous Solid dispersions have generated much interest from pharma-
state and crystalline state of drugs in solid dispersions. However, ceutical scientists because of the increasing number of drug candi-
the crystallinities under 5–10% fraction may not be detected by dates which is poorly water soluble and the recent advances on
PXRD [23]. this area. Although solid dispersions have been investigated for
such a long time, some novel carriers, additives and new prepara-
8.3. Fourier Transformed Infrared spectroscopy (FTIR) tion, characterization techniques have just been applied in recent
years. This brings new hope to develop more solid dispersion prod-
FTIR is a common technique used to investigate the intermolec- ucts in the future. Recent advances on solid dispersion area can be
ular interaction and drug–carrier compatibility because it can divided into four main issues: (i) applying new carriers, (ii) adding
detect the physical and chemical reaction between drug and car- new additives such as surfactants, superdisintegrants and pH mod-
rier. Hydrogen bonding between drugs and carriers which is very ifiers, (iii) developing novel preparation and characterization
important to explain the physical state and the stability of drugs methods, (iv) elucidating the thermodynamic mechanism of many
in solid dispersions can also be identified by FTIR. Miyazaki et al. processes in the preparation, formulation, dissolution and storage
[39] studied the crystallization rates of nitrendipine (NTR) enanti- stage. These issues are interrelated and will be continuously inves-
omers in the presence of HPMC, HPMCP and PVP. FTIR results indi- tigated in the coming time.
cated that PVP interacted with NTR through hydrogen bonding at To broaden and optimize the various carriers used in solid dis-
the NH moiety of NTR, and almost the same degrees of shift in persions, several studies have introduced new carriers while many
wavenumber for NH stretching suggested a similar strength of other studies utilize more than two polymers as carriers to com-
hydrogen bonding interaction for enantiomers: ()NTR and bine the advantages of each polymer. Some novel carriers used in
(+)NTR. Therefore, similar degree of physical stability between recent years are soluplusÒ, inutec SP1 or KollicoatÒ IR. Lin et al.
()NTR and (+)NTR was observed in PVP based solid dispersions [149] synthesized nano-sized flake carboxymethyl cassava starch
whereas the overall crystallization rate and the nucleation rate of (CMCS) to prepare solid dispersions loading acetylsalicylic acid.
(+)-NTR were lower than those of ()NTR in HPMC or HPMCP The results suggested that nano-sized CMCS based solid disper-
based solid dispersions because of stereoselective interaction sions had much better dispersion capability for the drug than the
between enantiomers and these carriers. The hydrogen bonding normal CMCS and micro-sized CMCS based solid dispersions. The
between PVP and NTR also explained for the lower growth rate dissolution rate of nano-sized CMCS based solid dispersions was
of NTR crystal in PVP based solid dispersions compared to HPMC considerably higher than that of pure drug. Al-Obaidi et al. [150]
and HPMCP based solid dispersions. investigated the impact of the third polymer on the physical stabil-
ity of drugs in binary solid dispersions. Poly[N-(2-hydroxypro-
8.4. Thermal Gravimetry Analysis (TGA) pyl)methacrylate] (PHPMA) was added to drug–PVP solid
dispersions because it can form hydrogen bonds with drugs (gris-
TGA is a method of thermal analysis that measures the weight eofulvin, progesterone, phenindione) while PVP cannot. The stabil-
change as a function of time and temperature, thereby providing ity results showed that the ternary solid dispersions crystallized
information about the stability of a material and the compatibility more slowly than binary solid dispersions. It was concluded that
of different materials in a solid dispersion mixture. This method the incorporation of a bridging polymer which can form hydrogen
can provide useful information about the stability of drugs and bonds with the API and the second polymer can improve signifi-
carriers as well as the chemical and physical processes in solid dis- cantly the stability of amorphous drug in immiscible binary solid
persions to decide the preparation method and the processing dispersions.
parameters for solid dispersion preparation. Other common appli- Many novel additives are also added in solid dispersions to
cations in the pharmaceutical sciences include the determination increase the solubility and the stability of API. These additives in-
of moisture and solvent content as well as decomposition, vapori- clude novel surfactants, superdisintegrants, and pH modifiers
zation or sublimation temperatures [147]. However, this technique [151–153]. Szu } ts et al. [59] incorporated sucrose laurate into gem-
is not effective for materials that do not exhibit a weight change fibrozil-PEG6000 solid dispersions. The results showed that the
during degradation and some processes which do not involve the presence of the novel surfactant did not influence the solid-state
loss of mass. Similar to DSC method, TGA results are changeable characteristics of the API significantly. The dissolution of gemfibro-
and depend on the conditions of sample and experimental process zil from the ternary solid dispersion systems was markedly im-
which is difficult to compare the work of one researcher to another proved compared to the binary solid dispersion systems. It is
[147]. Frizon et al. [148] used TGA method to check the thermal important to notice that two-thirds of poorly water-soluble drugs
stability of loratadine in the temperature range of the preparation have pH-dependent solubility. Accordingly, pH modifiers can be
810 C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813

proposed as a promising and important approach to improve the completely elucidate the structure and dissolution mechanism of
solubility of such drugs in solid dispersions. Tran et al. [154] solid dispersions in the near future. This can lead to an optimal
proved that the alkalizers in PEG 6000 based solid dispersions syn- selection of carriers and preparation methods for each poorly
ergistically enhanced dissolution of telmisartan, a weak acid, not water-soluble drug and thus will overcome the bioavailability
only by modulating the micro-environmental pH (pHM), but also problems of drugs as well as the stability and scalability problems
by changing drug crystallinity to an amorphous form via molecular of solid dispersions.
interactions. Srinarong et al. [155] showed that the dissolution rate
was more increased when the superdisintegrant was incorporated
Acknowledgements
in solid dispersions than when it was physically mixed with the
solid dispersions to prepare tablets. The dissolution rate enhance-
This work was supported by the Korean Health Technology R&D
ment which was explained by the prevention of drug recrystalliza-
Project, Ministry for Health and Welfare (A092018).
tion during dissolution process was strongly dependent on the
type of superdisintegrants.
KinetiSolÒ Dispersing (KSD) is a novel method of solid disper- References
sion preparation. It is a high energy mixing process for the produc-
tion of amorphous pharmaceutical solids which uses a series of [1] J.A. Baird, L.S. Taylor, Evaluation of amorphous solid dispersion properties
using thermal analysis techniques, Adv. Drug Delivery Rev. 64 (2012) 396–
rapidly rotating blades to process the drug and polymeric carrier 421.
through a combination of kinetic and thermal energy without the [2] C.A. Lipinski, F. Lombardo, B.W. Dominy, P.J. Feeney, Experimental and
aid of external heating sources [156]. Dinunzio et al. [156] computational approaches to estimate solubility and permeability in drug
discovery and development settings, Adv. Drug Delivery Rev. 46 (2012) 3–26.
prepared solid dispersions of itraconazole and hypromellose by [3] G. Van den Mooter, The use of amorphous solid dispersions: a formulation
KSD and compared to identical formulations produced by hot melt strategy to overcome poor solubility and dissolution rate, Drug Discovery
extrusion. Results showed that amorphous API was formed within Today Technol. 9 (2011) 79–85.
[4] FDA, Waiver of In Vivo Bioavailability and Bioequivalence Studies for
15 s by KSD while hot melt extrusion required over 300 s. The dis- Immediate-Release Solid Oral Dosage Forms based on a Biopharmaceutics
solution rates of compositions manufactured by KSD were also im- Classification System, Guidance for Industry, 2000.
proved compared to hot melt extrusion processed material. KSD [5] Y. Kawabata, K. Wada, M. Nakatani, S. Yamada, S. Onoue, Formulation design
for poorly water-soluble drugs based on biopharmaceutics classification
was considered a promising manufacturing technique for solid dis- system: basic approaches and practical applications, Int. J. Pharm. 420 (2011)
persion preparation which provided benefits over conventional 1–10.
techniques in enhancing mixing for improved homogeneity and [6] T. Vasconcelos, B. Sarmento, P. Costa, Solid dispersions as strategy to improve
oral bioavailability of poor water soluble drugs, Drug Discovery Today 12
reducing processing times. Various novel characterization tech-
(2007) 1068–1075.
niques have also been introduced to the area of solid dispersions [7] S.M. Berge, L.D. Bighley, D.C. Monkhouse, Pharmaceutical salts, J. Pharm. Sci.
and showed many advantages. High performance DSC (HDSC), Pro- 66 (1977) 1–19.
ject Rapid Heat/Cool (RHC) and chip calorimetry have been applied [8] J. Rautio, H. Kumpulainen, T. Heimbach, R. Oliyai, D. Oh, T. Järvinen, J.
Savolainen, Prodrugs: design and clinical applications, Nat. Rev. Drug
in solid dispersion area to overcome some shortages of DSC relat- Discovery 7 (2008) 255–270.
ing to slow cooling/heating scan which can cause sample degrada- [9] B.E. Rabinow, Nanosuspensions in drug delivery, Nat. Rev. Drug Discovery 3
tion and undesired transformation to a different solid state form (2004) 785–796.
[10] T. Loftsson, D. Duchene, Cyclodextrins and their pharmaceutical applications,
[1,157]. Some thermal techniques such as nanothermal analysis Int. J. Pharm. 329 (2007) 1–11.
(NTA), localized thermal analysis (LTA). . .can carry out local ther- [11] A. Lavasanifar, J. Samuel, G.S. Kwon, Poly (ethylene oxide)-block-poly (l-
mal analysis at defined points on a surface of solid dispersion amino acid) micelles for drug delivery, Adv. Drug Delivery Rev. 54 (2002)
169–190.
and map thermal properties during imaging to show drug–poly- [12] E.A. Mueller, J.M. Kovarik, J.B. van Bree, W. Tetzloff, J. Grevel, K. Kutz,
mer miscibility [1,158]. High resolution atomic force microscopy Improved dose linearity of cyclosporine pharmacokinetics from a
was also utilized for the investigation of the solid dispersion struc- microemulsion formulation, Pharm. Res. 11 (1994) 301–304.
[13] D.G. Fatouros, G.R. Deen, L. Arleth, B. Bergenstahl, F.S. Nielsen, J.S. Pedersen, A.
ture and properties, providing intuitively the spatial conformation, Mullertz, Structural development of self nano emulsifying drug delivery
the interactions between the API and carrier in solid dispersions at systems (SNEDDS) during in vitro lipid digestion monitored by small-angle X-
the molecular level [159]. This technique can even confirm directly ray scattering, Pharm. Res. 24 (2007) 1844–1853.
[14] R.H. Müller, K. Peters, Nanosuspensions for the formulation of poorly soluble
intermolecular hydrogen bonding within solid dispersions and was
drugs: I. Preparation by a size-reduction technique, Int. J. Pharm. 160 (1998)
used by Yu et al. [159] to investigate the structure and dissolution 229–237.
properties of a solid dispersion containing lansoprazole and PVP. [15] S.G. Potta, S. Minemi, R.K. Nukala, C. Peinado, D.A. Lamprou, A. Urquhart, D.
The development of characterization techniques will help to dis- Douroumis, Development of solid lipid nanoparticles for enhanced solubility
of poorly soluble drugs, J. Biomed. Nanotechnol. 6 (2010) 634–640.
cover the thermodynamic property and mechanism of solid disper- [16] W.L. Chiou, S. Riegelman, Pharmaceutical applications of solid dispersion
sions; therefore, more strategies will be applied to overcome the systems, J. Pharm. Sci. 60 (1971) 1281–1302.
remaining problems of solid dispersions in the future. [17] M. Vasanthavada, W.-Q. Tong, Y. Joshi, M.S. Kislalioglu, Phase behavior of
amorphous molecular dispersions I: determination of the degree and
mechanism of solid solubility, Pharm. Res. 21 (2004) 1598–1606.
[18] B. Chauhan, S. Shimpi, A. Paradkar, Preparation and evaluation of
10. Conclusions glibenclamide-polyglycolized glycerides solid dispersions with silicon
dioxide by spray drying technique, Eur. J. Pharm. Sci. 26 (2005) 219–230.
[19] V.B. Pokharkar, L.P. Mandpe, M.N. Padamwar, A.A. Ambike, K.R. Mahadik, A.
Solid dispersions are currently considered one of the most Paradkar, Development, characterization and stabilization of amorphous
effective methods to solve the low bioavailability problem of form of a low Tg drug, Powder Technol. 167 (2006) 20–25.
[20] G. Van den Mooter, I. Weuts, T. De Ridder, N. Blaton, Evaluation of Inutec SP1
poorly water-soluble drugs. Although some problems relating to as a new carrier in the formulation of solid dispersions for poorly soluble
instability and scalability still remain; novel and optimized manu- drugs, Int. J. Pharm. 316 (2006) 1–6.
facturing techniques with high potential to overcome these prob- [21] K. Sekiguchi, N. Obi, Studies on absorption of eutectic mixture. I. A
comparison of the behavior of eutectic mixture of sulfathiazole and that of
lems are being introduced, thanks to academic and industrial
ordinary sulfathiazole in man, Chem. Pharm. Bull. 9 (1961) 866–872.
researches. This review documents current efforts to overcome [22] D.Q. Craig, The mechanisms of drug release from solid dispersions in water-
these problems and discusses critical aspects for better under- soluble polymers, Int. J. Pharm. 231 (2002) 131–144.
standing of solid dispersions. The deeper investigation in physico- [23] C. Leuner, J. Dressman, Improving drug solubility for oral delivery using solid
dispersions, Eur. J. Pharm. Biopharm. 50 (2000) 47–60.
chemical properties and interactions of drugs and carriers as well [24] R. Jachowicz, Dissolution rates of partially water-soluble drugs from solid
as the improvement of characterization techniques may help to dispersion systems. I. Prednisolone, Int. J. Pharm. 35 (1987) 1–5.
C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813 811

[25] N. Zajc, A. Obreza, M. Bele, S. Srčič, Physical properties and dissolution [52] C. Stevens, A. Meriggi, M. Peristeropoulou, P. Christov, K. Booten, B. Levecke,
behaviour of nifedipine/mannitol solid dispersions prepared by hot melt A. Vandamme, N. Pittevils, T.F. Tadros, Polymeric surfactants based on inulin,
method, Int. J. Pharm. 291 (2005) 51–58. a polysaccharide extracted from chicory. 1. Synthesis and interfacial
[26] S. Okonogi, T. Oguchi, E. Yonemochi, S. Puttipipatkhachorn, K. Yamamoto, properties, Biomacromolecules 2 (2001) 1256–1259.
Improved dissolution of ofloxacin via solid dispersion, Int. J. Pharm. 156 [53] C.V. Stevens, A. Meriggi, K. Booten, Chemical modification of inulin, a valuable
(1997) 175–180. renewable resource, and its industrial applications, Biomacromolecules 2
[27] D. Van Drooge, W. Hinrichs, M. Visser, H. Frijlink, Characterization of the (2001) 1–16.
molecular distribution of drugs in glassy solid dispersions at the nano-meter [54] J. Moes, S. Koolen, A. Huitema, J. Schellens, J. Beijnen, B. Nuijen,
scale, using differential scanning calorimetry and gravimetric water vapour Pharmaceutical development and preliminary clinical testing of an oral
sorption techniques, Int. J. Pharm. 310 (2006) 220–229. solid dispersion formulation of docetaxel (ModraDoc001), Int. J. Pharm. 420
[28] N. Tanaka, K. Imai, K. Okimoto, S. Ueda, Y. Tokunaga, R. Ibuki, K. Higaki, T. (2011) 244–250.
Kimura, Development of novel sustained-release system, disintegration- [55] H.N. Joshi, R.W. Tejwani, M. Davidovich, V.P. Sahasrabudhe, M. Jemal, M.S.
controlled matrix tablet (DCMT) with solid dispersion granules of nilvadipine Bathala, S.A. Varia, A. Serajuddin, Bioavailability enhancement of a poorly
(II): in vivo evaluation, J. Controlled Release 112 (2006) 51–56. water-soluble drug by solid dispersion in polyethylene glycol–polysorbate 80
[29] T. Vilhelmsen, H. Eliasen, T. Schæfer, Effect of a melt agglomeration process on mixture, Int. J. Pharm. 269 (2004) 251–258.
agglomerates containing solid dispersions, Int. J. Pharm. 303 (2005) 132–142. [56] A.N. Ghebremeskel, C. Vemavarapu, M. Lodaya, Use of surfactants as
[30] N.A. Urbanetz, Stabilization of solid dispersions of nimodipine and plasticizers in preparing solid dispersions of poorly soluble API: selection of
polyethylene glycol 2000, Eur. J. Pharm. Sci. 28 (2006) 67–76. polymer–surfactant combinations using solubility parameters and testing the
[31] H. Chauhan, C. Hui-Gu, E. Atef, Correlating the behavior of polymers in processability, Int. J. Pharm. 328 (2007) 119–129.
solution as precipitation inhibitor to its amorphous stabilization ability in [57] C. Goddeeris, T. Willems, K. Houthoofd, J. Martens, G. Van den Mooter,
solid dispersions, J. Pharm. Sci. 102 (2013) 1924–1935. Dissolution enhancement of the anti-HIV drug UC 781 by formulation in a
[32] M.M. Crowley, F. Zhang, M.A. Repka, S. Thumma, S.B. Upadhye, S.K. Battu, J.W. ternary solid dispersion with TPGS 1000 and Eudragit E100, Eur. J. Pharm.
McGinity, C. Martin, Pharmaceutical applications of hot-melt extrusion: Part Biopharm. 70 (2008) 861–868.
I, Drug Dev. Ind. Pharm. 33 (2007) 909–926. [58] D.-H. Won, M.-S. Kim, S. Lee, J.-S. Park, S.-J. Hwang, Improved
[33] L.S. Taylor, G. Zografi, Spectroscopic characterization of interactions between physicochemical characteristics of felodipine solid dispersion particles by
PVP and indomethacin in amorphous molecular dispersions, Pharm. Res. 14 supercritical anti-solvent precipitation process, Int. J. Pharm. 301 (2005) 199–
(1997) 1691–1698. 208.
[34] H. Bley, B. Fussnegger, R. Bodmeier, Characterization and stability of solid [59] A. Szu} ts, P. Láng, R. Ambrus, L. Kiss, M.A. Deli, P. Szabó-Révész, Applicability of
dispersions based on PEG/polymer blends, Int. J. Pharm. 390 (2010) 165–173. sucrose laurate as surfactant in solid dispersions prepared by melt
[35] S.-C. Shin, I.-J. Oh, Y.-B. Lee, H.-K. Choi, J.-S. Choi, Enhanced dissolution of technology, Int. J. Pharm. 410 (2011) 107–110.
furosemide by coprecipitating or cogrinding with crospovidone, Int. J. Pharm. [60] H. De Waard, W. Hinrichs, M. Visser, C. Bologna, H. Frijlink, Unexpected
175 (1998) 17–24. differences in dissolution behavior of tablets prepared from solid dispersions
[36] J. Huang, R.J. Wigent, C.M. Bentzley, J.B. Schwartz, Nifedipine solid dispersion with a surfactant physically mixed or incorporated, Int. J. Pharm. 349 (2008)
in microparticles of ammonio methacrylate copolymer and ethylcellulose 66–73.
binary blend for controlled drug delivery: effect of drug loading on release [61] J. Huang, R.J. Wigent, J.B. Schwartz, Nifedipine molecular dispersion in
kinetics, Int. J. Pharm. 319 (2006) 44–54. microparticles of ammonio methacrylate copolymer and ethylcellulose
[37] H.-T. Lim, P. Balakrishnan, D.H. Oh, K.H. Joe, Y.R. Kim, D.H. Hwang, Y.-B. Lee, binary blends for controlled drug delivery: effect of matrix composition,
C.S. Yong, H.-G. Choi, Development of novel sibutramine base-loaded solid Drug Dev. Ind. Pharm. 32 (2006) 1185–1197.
dispersion with gelatin and HPMC: physicochemical characterization and [62] J. Desai, K. Alexander, A. Riga, Characterization of polymeric dispersions of
pharmacokinetics in beagle dogs, Int. J. Pharm. 397 (2010) 225–230. dimenhydrinate in ethyl cellulose for controlled release, Int. J. Pharm. 308
[38] J.J. García-Rodriguez, P.M. de la Torre-Iglesias, M.C. Vegas-Sánchez, S. (2006) 115–123.
Torrado-Durán, F. Bolás-Fernández, S. Torrado-Santiago, Changed [63] Z. Iqbal, A. Babar, M. Ashraf, Controlled-release naproxen using micronized
crystallinity of mebendazole solid dispersion: improved anthelmintic ethyl cellulose by wet-granulation and solid-dispersion method, Drug Dev.
activity, Int. J. Pharm. 403 (2011) 23–28. Ind. Pharm. 28 (2002) 129–134.
[39] T. Miyazaki, Y. Aso, S. Yoshioka, T. Kawanishi, Differences in crystallization [64] F. Cui, M. Yang, Y. Jiang, D. Cun, W. Lin, Y. Fan, Y. Kawashima, Design of
rate of nitrendipine enantiomers in amorphous solid dispersions with HPMC sustained-release nitrendipine microspheres having solid dispersion
and HPMCP, Int. J. Pharm. 407 (2011) 111–118. structure by quasi-emulsion solvent diffusion method, J. Controlled Release
[40] M. Zhang, H. Li, B. Lang, K. O’Donnell, H. Zhang, Z. Wang, Y. Dong, C. Wu, R.O. 91 (2003) 375–384.
Williams III, Formulation and delivery of improved amorphous fenofibrate [65] T. Ozeki, H. Yuasa, Y. Kanaya, Controlled release from solid dispersion
solid dispersions prepared by thin film freezing, Eur. J. Pharm. Biopharm. 82 composed of poly (ethylene oxide)–CarbopolÒ interpolymer complex with
(2012) 534–544. various cross-linking degrees of CarbopolÒ, J. Controlled Release 63 (2000)
[41] S. Bialleck, H. Rein, Preparation of starch-based pellets by hot-melt extrusion, 287–295.
Eur. J. Pharm. Biopharm. 79 (2011) 440–448. [66] T. Ohara, S. Kitamura, T. Kitagawa, K. Terada, Dissolution mechanism of
[42] D. Van Drooge, W. Hinrichs, H. Frijlink, Anomalous dissolution behaviour of poorly water-soluble drug from extended release solid dispersion system
tablets prepared from sugar glass-based solid dispersions, J. Controlled with ethylcellulose and hydroxypropylmethylcellulose, Int. J. Pharm. 302
Release 97 (2004) 441–452. (2005) 95–102.
[43] J.C. Shah, J.R. Chen, D. Chow, Preformulation study of etoposide: II. Increased [67] T.T.-D. Tran, P.H.-L. Tran, J. Lim, J.B. Park, S.-K. Choi, B.-J. Lee, Physicochemical
solubility and dissolution rate by solid–solid dispersions, Int. J. Pharm. 113 principles of controlled release solid dispersion containing a poorly water-
(1995) 103–111. soluble drug, Ther. Delivery 1 (2010) 51–62.
[44] E. Karavas, E. Georgarakis, M.P. Sigalas, K. Avgoustakis, D. Bikiaris, [68] N. Kolašinac, K. Kachrimanis, I. Homšek, B. Grujić, Z. Ðurić, S. Ibrić, Solubility
Investigation of the release mechanism of a sparingly water-soluble drug enhancement of desloratadine by solid dispersion in poloxamers, Int. J.
from solid dispersions in hydrophilic carriers based on physical state of drug, Pharm. 436 (2012) 161–170.
particle size distribution and drug–polymer interactions, Eur. J. Pharm. [69] G. Muhrer, U. Meier, F. Fusaro, S. Albano, M. Mazzotti, Use of compressed gas
Biopharm. 66 (2007) 334–347. precipitation to enhance the dissolution behavior of a poorly water-soluble
[45] M.R. Visser, L. Baert, G.v.t. Klooster, L. Schueller, M. Geldof, I. drug: generation of drug microparticles and drug–polymer solid dispersions,
Vanwelkenhuysen, H. de Kock, S. De Meyer, H.W. Frijlink, J. Rosier, Inulin Int. J. Pharm. 308 (2006) 69–83.
solid dispersion technology to improve the absorption of the BCS Class IV [70] E. Karavas, G. Ktistis, A. Xenakis, E. Georgarakis, Effect of hydrogen bonding
drug TMC240, Eur. J. Pharm. Biopharm. 74 (2010) 233–238. interactions on the release mechanism of felodipine from nanodispersions
[46] N. Passerini, B. Albertini, M.L. González-Rodrı´guez, C. Cavallari, L. Rodriguez, with polyvinylpyrrolidone, Eur. J. Pharm. Biopharm. 63 (2006) 103–114.
Preparation and characterisation of ibuprofen–poloxamer 188 granules [71] C.W. Pouton, Formulation of poorly water-soluble drugs for oral
obtained by melt granulation, Eur. J. Pharm. Sci. 15 (2002) 71–78. administration: physicochemical and physiological issues and the lipid
[47] S. Jagdale, S. Patil, B. Kuchekar, A. Chabukswar, Preparation and formulation classification system, Eur. J. Pharm. Sci. 29 (2006) 278–287.
characterization of metformin hydrochloride—Compritol 888 ATO solid [72] J.P. Moschwitzer, Drug nanocrystals in the commercial pharmaceutical
dispersion, J. Young Pharm. 3 (2011) 197. development process, Int. J. Pharm. (2012).
[48] F. Damian, N. Blaton, L. Naesens, J. Balzarini, R. Kinget, P. Augustijns, G. Van [73] A. Serajuddin, Solid dispersion of poorly water-soluble drugs: early promises,
den Mooter, Physicochemical characterization of solid dispersions of the subsequent problems, and recent breakthroughs, J. Pharm. Sci. 88 (1999)
antiviral agent UC-781 with polyethylene glycol 6000 and Gelucire 44/14, 1058–1066.
Eur. J. Pharm. Sci. 10 (2000) 311–322. [74] R. Ghaderi, P. Artursson, J. Carlfors, Preparation of biodegradable
[49] A. Kalivoda, M. Fischbach, P. Kleinebudde, Application of mixtures of microparticles using solution-enhanced dispersion by supercritical fluids
polymeric carriers for dissolution enhancement of oxeglitazar using hot- (SEDS), Pharm. Res. 16 (1999) 676–681.
melt extrusion, Int. J. Pharm. 439 (2012) 145–156. [75] S. Bhattacharya, R. Suryanarayanan, Local mobility in amorphous
[50] S. Shah, S. Maddineni, J. Lu, M.A. Repka, Melt extrusion with poorly soluble pharmaceuticals—characterization and implications on stability, J. Pharm.
drugs, Int. J. Pharm. (2012). Sci. 98 (2009) 2935–2953.
[51] W. Ali, A.C. Williams, C.F. Rawlinson, Stochiometrically governed molecular [76] M. Yoshioka, B.C. Hancock, G. Zografi, Crystallization of indomethacin from
interactions in drug: poloxamer solid dispersions, Int. J. Pharm. 391 (2010) the amorphous state below and above its glass transition temperature, J.
162–168. Pharm. Sci. 83 (1994) 1700–1705.
812 C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813

[77] S. Vyazovkin, I. Dranca, Physical stability and relaxation of amorphous [104] M.J. Rathbone, J. Hadgraft, M.S. Roberts, Modified-Release Drug Delivery
indomethacin, J. Phys. Chem. B 109 (2005) 18637–18644. Technology, first ed., Marcel Dekker, New York, 2003. pp. 126–128.
[78] C. Bhugra, R. Shmeis, S.L. Krill, M.J. Pikal, Prediction of onset of crystallization [105] J. Ford, M. Rubinstein, Formulation and ageing of tablets prepared from
from experimental relaxation times. II. Comparison between predicted and indomethacin-polyethylene glycol 6000 solid dispersions, Pharm. Acta Helv.
experimental onset times, J. Pharm. Sci. 97 (2008) 455–472. 55 (1980) 1.
[79] S.P. Duddu, T.D. Sokoloski, Dielectric analysis in the characterization of [106] A. Seo, P. Holm, H.G. Kristensen, T. Schæfer, The preparation of agglomerates
amorphous pharmaceutical solids. 1. Molecular mobility in poly containing solid dispersions of diazepam by melt agglomeration in a high
(vinylpyrrolidone)–water systems in the glassy state, J. Pharm. Sci. 84 shear mixer, Int. J. Pharm. 259 (2003) 161–171.
(1995) 773–776. [107] M.K. Gupta, Y.-C. Tseng, D. Goldman, R.H. Bogner, Hydrogen bonding with
[80] B.C. Hancock, G. Zografi, The relationship between the glass transition adsorbent during storage governs drug dissolution from solid-dispersion
temperature and the water content of amorphous pharmaceutical solids, granules, Pharm. Res. 19 (2002) 1663–1672.
Pharm. Res. 11 (1994) 471–477. [108] N.G. Sahoo, M. Kakran, L. Li, Z. Judeh, R.H. Müller, Dissolution enhancement of
[81] Z. Ayenew, A. Paudel, G. Van den Mooter, Can compression induce demixing a poorly water-soluble antimalarial drug by means of a modified multi-fluid
in amorphous solid dispersions? A case study of naproxen–PVP K25, Eur. J. nozzle pilot spray drier, Mater. Sci. Eng., C: Mater. Biol. Appl. 31 (2011) 391–
Pharm. Biopharm. 81 (2012) 207–213. 399.
[82] K. Six, T. Daems, J. de Hoon, A. Van Hecken, M. Depre, M.-P. Bouche, P. Prinsen, [109] T. Hoshino, F. Kusaki, I. Fukui, Solid Dispersion Preparation, European Patent
G. Verreck, J. Peeters, M.E. Brewster, Guy Van den Mooter, Clinical study of EP 1847260 A3 (2007).
solid dispersions of itraconazole prepared by hot-stage extrusion, Eur. J. [110] V. Kumar, Solid Dispersions of Opioid Antagonists, U.S. Patent 7,914,776
Pharm. Sci. 24 (2005) 179–186. (2011).
[83] M. Vasanthavada, W.-Q. Tong, Y. Joshi, M.S. Kislalioglu, Phase behavior of [111] X. Wang, A. Michoel, G. Van den Mooter, Solid state characteristics of ternary
amorphous molecular dispersions II: role of hydrogen bonding in solid solid dispersions composed of PVP VA64, Myrj 52 and itraconazole, Int. J.
solubility and phase separation kinetics, Pharm. Res. 22 (2005) 440–448. Pharm. 303 (2005) 54–61.
[84] Y.-D. Yan, J.H. Sung, K.K. Kim, C.S. Yong, H.-G. Choi, Novel valsartan-loaded [112] A. Ceballos, M. Cirri, F. Maestrelli, G. Corti, P. Mura, Influence of formulation
solid dispersion with enhanced bioavailability and no crystalline changes, Int. and process variables on in vitro release of theophylline from directly-
J. Pharm. 422 (2011) 202–210. compressed Eudragit matrix tablets, Farmaco 60 (2005) 913–918.
[85] P. Mura, J.R. Moyano, M.L. Gonzalez-Rodriguez, A.M. Rabasco-Alvarez, M. [113] C. Li, C. Li, Y. Le, J.-F. Chen, Formation of bicalutamide nanodispersion for
Cirri, F. Maestrelli, Characterization and dissolution properties of ketoprofen dissolution rate enhancement, Int. J. Pharm. 404 (2011) 257–263.
in binary and ternary solid dispersions with polyethylene glycol and [114] K.A. Overhoff, J.D. Engstrom, B. Chen, B.D. Scherzer, T.E. Milner, K.P. Johnston,
surfactants, Drug Dev. Ind. Pharm. 31 (2005) 425–434. R.O. Williams, Novel ultra-rapid freezing particle engineering process for
[86] W.L. Chiou, S. Riegelman, Preparation and dissolution characteristics of enhancement of dissolution rates of poorly water-soluble drugs, Eur. J.
several fast-release solid dispersions of griseofulvin, J. Pharm. Sci. 58 (1969) Pharm. Biopharm. 65 (2007) 57–67.
1505–1510. [115] A. Paudel, Z.A. Worku, J. Meeus, S. Guns, G. Van den Mooter, Manufacturing of
[87] F.-Q. Li, J.-H. Hu, J.-X. Deng, H. Su, S. Xu, J.-Y. Liu, In vitro controlled release of solid dispersions of poorly water soluble drugs by spray drying: formulation
sodium ferulate from Compritol 888 ATO-based matrix tablets, Int. J. Pharm. and process considerations, Int. J. Pharm. (2012).
324 (2006) 152–157. [116] P. Srinarong, H. de Waard, H.W. Frijlink, W.L. Hinrichs, Improved dissolution
[88] W.-W. Yao, T.-C. Bai, J.-P. Sun, C.-W. Zhu, J. Hu, H.-L. Zhang, Thermodynamic behavior of lipophilic drugs by solid dispersions: the production process as
properties for the system of silybin and poly (ethylene glycol) 6000, starting point for formulation considerations, Expert Opin. Drug Delivery 8
Thermochim. Acta 437 (2005) 17–20. (2011) 1121–1140.
[89] P. Barmpalexis, K. Kachrimanis, E. Georgarakis, Solid dispersions in the [117] D.N. Bikiaris, Solid dispersions, Part I: recent evolutions and future
development of a nimodipine floating tablet formulation and optimization by opportunities in manufacturing methods for dissolution rate enhancement
artificial neural networks and genetic programming, Eur. J. Pharm. Biopharm. of poorly water-soluble drugs, Expert Opin. Drug Delivery 8 (2011) 1501–
77 (2011) 122–131. 1519.
[90] B. Albertini, M. Mezzena, N. Passerini, L. Rodriguez, S. Scalia, Evaluation of [118] P.G. Dan Smithey, Lynne Taylor, Amorphous solid dispersions: an enabling
spray congealing as technique for the preparation of highly loaded solid lipid formulation technology for oral delivery of poorly water soluble drugs, AAPS
microparticles containing the sunscreen agent, avobenzone, J. Pharm. Sci. 98 Newsmag. 16 (2013) 11–14.
(2009) 2759–2769. [119] D.J. van Drooge, K. Braeckmans, W.L. Hinrichs, K. Remaut, S.C. De Smedt, H.W.
[91] N. Passerini, S. Qi, B. Albertini, M. Grassi, L. Rodriguez, D.Q. Craig, Solid lipid Frijlink, Characterization of the mode of incorporation of lipophilic
microparticles produced by spray congealing: influence of the atomizer on compounds in solid dispersions at the nanoscale using fluorescence
microparticle characteristics and mathematical modeling of the drug release, resonance energy transfer (FRET), Macromol. Rapid Commun. 27 (2006)
J. Pharm. Sci. 99 (2010) 916–931. 1149–1155.
[92] N. Passerini, B. Albertini, B. Perissutti, L. Rodriguez, Evaluation of melt [120] T. Purvis, M.E. Mattucci, M.T. Crisp, K.P. Johnston, R.O. Williams, Rapidly
granulation and ultrasonic spray congealing as techniques to enhance the dissolving repaglinide powders produced by the ultra-rapid freezing process,
dissolution of praziquantel, Int. J. Pharm. 318 (2006) 92–102. AAPS PharmSciTech 8 (2007) 52–60.
[93] A. Fini, L. Rodriguez, C. Cavallari, B. Albertini, N. Passerini, Ultrasound- [121] J. Hu, T.L. Rogers, J. Brown, T. Young, K.P. Johnston, R.O. Williams Iii,
compacted and spray-congealed indomethacin/polyethyleneglycol systems, Improvement of dissolution rates of poorly water soluble APIs using novel
Int. J. Pharm. 247 (2002) 11–22. spray freezing into liquid technology, Pharm. Res. 19 (2002) 1278–1284.
[94] M.M. Mehanna, A.M. Motawaa, M.W. Samaha, In sight into tadalafil–block [122] J. Hu, K.P. Johnston, R. Williams 3rd, Spray freezing into liquid (SFL) particle
copolymer binary solid dispersion: mechanistic investigation of dissolution engineering technology to enhance dissolution of poorly water soluble drugs:
enhancement, Int. J. Pharm. 402 (2010) 78–88. organic solvent versus organic/aqueous co-solvent systems, Eur. J. Pharm. Sci.
[95] E. Verhoeven, T. De Beer, E. Schacht, G. Van den Mooter, J.P. Remon, C. 20 (2003) 295.
Vervaet, Influence of polyethylene glycol/polyethylene oxide on the release [123] H.H. Tong, Z. Du, G.N. Wang, H. Chan, Q. Chang, L. Lai, A.H. Chow, Y. Zheng,
characteristics of sustained-release ethylcellulose mini-matrices produced by Spray freeze drying with polyvinylpyrrolidone and sodium caprate for
hot-melt extrusion: in vitro and in vivo evaluations, Eur. J. Pharm. Biopharm. improved dissolution and oral bioavailability of oleanolic acid, a BCS Class
72 (2009) 463–470. IV compound, Int. J. Pharm. 404 (2011) 148–158.
[96] M. Maniruzzaman, J.S. Boateng, M. Bonnefille, A. Aranyos, J.C. Mitchell, D. [124] D.-J. van Drooge, W.L. Hinrichs, B.H. Dickhoff, M.N. Elli, M.R. Visser, G.S.
Douroumis, Taste masking of paracetamol by hot-melt extrusion: an in vitro Zijlstra, H.W. Frijlink, Spray freeze drying to produce a stable-
and in vivo evaluation, Eur. J. Pharm. Biopharm. 80 (2012) 433–442. tetrahydrocannabinol containing inulin-based solid dispersion powder
[97] J. Moes, S. Koolen, A. Huitema, J. Schellens, J. Beijnen, B. Nuijen, Development suitable for inhalation, Eur. J. Pharm. Sci. 26 (2005) 231–240.
of an oral solid dispersion formulation for use in low-dose metronomic [125] K.A. Overhoff, A. Moreno, D.A. Miller, K.P. Johnston, R.O. Williams, Solid
chemotherapy of paclitaxel, Eur. J. Pharm. Biopharm. 83 (2013) 87–94. dispersions of itraconazole and enteric polymers made by ultra-rapid
[98] L. Saerens, L. Dierickx, B. Lenain, C. Vervaet, J.P. Remon, T.D. Beer, Raman freezing, Int. J. Pharm. 336 (2007) 122–132.
spectroscopy for the in-line polymer–drug quantification and solid state [126] L. Yu, Amorphous pharmaceutical solids: preparation, characterization and
characterization during a pharmaceutical hot-melt extrusion process, Eur. J. stabilization, Adv. Drug Delivery Rev. 48 (2001) 27–42.
Pharm. Biopharm. 77 (2011) 158–163. [127] E. Badens, V. Majerik, G. Horváth, L. Szokonya, N. Bosc, E. Teillaud, G. Charbit,
[99] M. Williams, Y. Tian, D.S. Jones, G.P. Andrews, Hot-melt extrusion technology: Comparison of solid dispersions produced by supercritical antisolvent and
optimizing drug delivery, Eur. J. Parenteral Sci. Pharm. Sci. 15 (2010) 61. spray-freezing technologies, Int. J. Pharm. 377 (2009) 25–34.
[100] J. Breitenbach, Melt extrusion: from process to drug delivery technology, Eur. [128] N. Shah, R.M. Iyer, H.J. Mair, D.S. Choi, H. Tian, R. Diodone, K. Fahnrich, A.
J. Pharm. Biopharm. 54 (2002) 107–117. Pabst-Ravot, K. Tang, E. Scheubel, J.F. Grippo, S.A. Moreira, Z. Go, J.
[101] H. He, R. Yang, X. Tang, In vitro and in vivo evaluation of fenofibrate solid Mouskountakis, T. Louie, P.N. Ibrahim, H. Sandhu, L. Rubia, H. Chokshi, D.
dispersion prepared by hot-melt extrusion, Drug Dev. Ind. Pharm. 36 (2010) Singhal, W. Malick, Improved human bioavailability of vemurafenib, a
681–687. practically insoluble drug, using an amorphous polymer-stabilized solid
[102] M. Maniruzzaman, J.S. Boateng, M.J. Snowden, D. Douroumis, A review of hot- dispersion prepared by a solvent-controlled coprecipitation process, J. Pharm.
melt extrusion: process technology to pharmaceutical products, ISRN Pharm. Sci. 102 (2013) 967–981.
2012 (2012) 436763. [129] N. Shah, H. Sandhu, W. Phuapradit, R. Pinal, R. Iyer, A. Albano, A. Chatterji, S.
[103] R. Chokshi, H. Zia, Hot-melt extrusion technique: a review, Iranian J. Pharm. Anand, D.S. Choi, K. Tang, H. Tian, H. Chokshi, D. Singhal, W. Malick,
Res. 3 (2010) 3–16. Development of novel microprecipitated bulk powder (MBP) technology for
C.Le-Ngoc Vo et al. / European Journal of Pharmaceutics and Biopharmaceutics 85 (2013) 799–813 813

manufacturing stable amorphous formulations of poorly soluble drugs, Int. J. [146] S. Guinot, F. Leveiller, The use of MTDSC to assess the amorphous phase
Pharm. 438 (2012) 53–60. content of a micronised drug substance, Int. J. Pharm. 192 (1999) 63–75.
[130] G. Sertsou, J. Butler, A. Scott, J. Hempenstall, T. Rades, Factors affecting [147] S.P. Stodghill, THERMAL ANALYSIS: thermal analysis—a review of techniques
incorporation of drug into solid solution with HPMCP during solvent change and applications in the pharmaceutical sciences, Am. Pharm. Rev. 13 (2010)
co-precipitation, Int. J. Pharm. 245 (2002) 99–108. 29.
[131] European Medicines Agency, Zelboraf {vemurafenib} CHMP Assessment [148] F. Frizon, J.d.O. Eloy, C.M. Donaduzzi, M.L. Mitsui, J.M. Marchetti, Dissolution
Report, Procedure No.: EMEA/H/C/002409, 2012, pp. 11–13. rate enhancement of loratadine in polyvinylpyrrolidone K-30 solid
[132] Z. Dong, A. Chatterji, H. Sandhu, D.S. Choi, H. Chokshi, N. Shah, Evaluation of dispersions by solvent methods, Powder Technol. 235 (2013) 532–539.
solid state properties of solid dispersions prepared by hot-melt extrusion and [149] X. Lin, W. Gao, C. Li, J. Chen, C. Yang, H. Wu, Nano-sized flake carboxymethyl
solvent co-precipitation, Int. J. Pharm. 355 (2008) 141–149. cassava starch as excipient for solid dispersions, Int. J. Pharm. 423 (2011)
[133] S. Hur, W.D. Kim, The electrospinning process and mechanical properties of 435–439.
nanofiber mats under vacuum conditions, Key Eng. Mater. 326 (2006) 393– [150] H. Al-Obaidi, P. Ke, S. Brocchini, G. Buckton, Characterization and stability of
396. ternary solid dispersions with PVP and PHPMA, Int. J. Pharm. 419 (2011) 20–
[134] G. Verreck, I. Chun, J. Rosenblatt, J. Peeters, A.V. Dijck, J. Mensch, M. Noppe, 27.
M.E. Brewster, Incorporation of drugs in an amorphous state into electrospun [151] H.-L.T. Phuong, T.T.-D. Tran, S.A. Lee, V.H. Nho, S.-C. Chi, B.-J. Lee, Roles of
nanofibers composed of a water-insoluble, nonbiodegradable polymer, J. MgO release from polyethylene glycol 6000-based solid dispersions on
Controlled Release 92 (2003) 349–360. microenvironmental pH, enhanced dissolution and reduced gastrointestinal
[135] G. Verreck, I. Chun, J. Peeters, J. Rosenblatt, M.E. Brewster, Preparation and damage of telmisartan, Arch. Pharm. Res. 34 (2011) 747–755.
characterization of nanofibers containing amorphous drug dispersions [152] M.-Y. Heo, Z.-Z. Piao, T.-W. Kim, Q.-R. Cao, A. Kim, B.-J. Lee, Effect of
generated by electrostatic spinning, Pharm. Res. 20 (2003) 810–817. solubilizing and microemulsifying excipients in polyethylene glycol 6000
[136] D.G. Yu, X.X. Shen, C. Branford-White, K. White, L.M. Zhu, S.W. Bligh, Oral solid dispersion on enhanced dissolution and bioavailability of ketoconazole,
fast-dissolving drug delivery membranes prepared from electrospun Arch. Pharm. Res. 28 (2005) 604–611.
polyvinylpyrrolidone ultrafine fibers, Nanotechnology 20 (2009) 055104. [153] P.H.-L. Tran, T.T.-D. Tran, Z.Z. Piao, T. Van Vo, J.B. Park, J. Lim, K.T. Oh, Y.-S.
[137] K. Kawakami, Miscibility analysis of particulate solid dispersions prepared by Rhee, B.-J. Lee, Physical properties and in vivo bioavailability in human
electrospray deposition, Int. J. Pharm. 433 (2012) 71–78. volunteers of isradipine using controlled release matrix tablet containing
[138] D.G. Yu, J.M. Yang, C. Branford-White, P. Lu, L. Zhang, L.M. Zhu, Third self-emulsifying solid dispersion, Int. J. Pharm. (2013).
generation solid dispersions of ferulic acid in electrospun composite [154] P.H.L. Tran, H.T.T. Tran, B.-J. Lee, Modulation of microenvironmental pH and
nanofibers, Int. J. Pharm. 400 (2010) 158–164. crystallinity of ionizable telmisartan using alkalizers in solid dispersions for
[139] X. Zhang, D. Wu, J. Lai, Y. Lu, Z. Yin, W. Wu, Piroxicam/2-hydroxypropyl-b- controlled release, J. Controlled Release 129 (2008) 59–65.
cyclodextrin inclusion complex prepared by a new fluid-bed coating [155] P. Srinarong, J. Faber, M. Visser, W. Hinrichs, H. Frijlink, Strongly enhanced
technique, J. Pharm. Sci. 98 (2009) 665–675. dissolution rate of fenofibrate solid dispersion tablets by incorporation of
[140] N. Sun, X. Wei, B. Wu, J. Chen, Y. Lu, W. Wu, Enhanced dissolution of superdisintegrants, Eur. J. Pharm. Biopharm. 73 (2009) 154–161.
silymarin/polyvinylpyrrolidone solid dispersion pellets prepared by a one- [156] J.C. DiNunzio, C. Brough, D.A. Miller, R.O. Williams III, J.W. McGinity, Fusion
step fluid-bed coating technique, Powder Technol. 182 (2008) 72–80. processing of itraconazole solid dispersions by KinetiSolÒ dispersing: a
[141] Y. Lu, X. Zhang, J. Lai, Z. Yin, W. Wu, Physical characterization of meloxicam- comparative study to hot melt extrusion, J. Pharm. Sci. 99 (2010) 1239–1253.
b-cyclodextrin inclusion complex pellets prepared by a fluid-bed coating [157] S. Guns, P. Kayaert, J.A. Martens, J. Van Humbeeck, V. Mathot, T. Pijpers, E.
method, Particuology 7 (2009) 1–8. Zhuravlev, C. Schick, G. Van den Mooter, Characterization of the copolymer
[142] N. Sun, X. Zhang, Y. Lu, W. Wu, In vitro evaluation and pharmacokinetics in poly (ethyleneglycol-g-vinylalcohol) as a potential carrier in the formulation
dogs of solid dispersion pellets containing Silybum marianum extract of solid dispersions, Eur. J. Pharm. Biopharm. 74 (2010) 239–247.
prepared by fluid-bed coating, Planta Med. 74 (2008) 126. [158] J. Zhang, M. Bunker, A. Parker, C.E. Madden-Smith, N. Patel, C.J. Roberts, The
[143] B. Shah, V.K. Kakumanu, A.K. Bansal, Analytical techniques for quantification stability of solid dispersions of felodipine in polyvinylpyrrolidone
of amorphous/crystalline phases in pharmaceutical solids, J. Pharm. Sci. 95 characterized by nanothermal analysis, Int. J. Pharm. 414 (2011) 210–217.
(2006) 1641–1665. [159] M. Yu, L. Sun, W. Li, Z. Lan, B. Li, L. Tan, M. Li, X. Yang, Investigation of
[144] G. Berndl, M. Degenhardt, M. Mäegerlein, G. Dispersyn, Itraconazole structure and dissolution properties of a solid dispersion of lansoprazole in
Compositions with Improved Bioavailability, U.S. Patent 2009/0214,656 polyvinylpyrrolidone, J. Mol. Struct. 1005 (2011) 70–77.
(2006). [160] N. Kohri, Y. Yamayoshi, H. Xin, K. Iseki, N. Sato, S. Todo, K. Miyazaki,
[145] I. Ghosh, J. Snyder, R. Vippagunta, M. Alvine, R. Vakil, W.Q. Tong, S. Improving the oral bioavailability of albendazole in rabbits by the solid
Vippagunta, Comparison of HPMC based polymers performance as carriers dispersion technique, Journal of Pharmacy and Pharmacology. 51 (2) (1999)
for manufacture of solid dispersions using the melt extruder, Int. J. Pharm. 159–164.
419 (2011) 12–19.

S-ar putea să vă placă și