Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

REVIEW

Amorphous Solid Dispersions: Utilization and Challenges in Drug


Discovery and Development
YAN HE, CHRIS HO

Pre-Development Sciences, LGCR, Waltham, Massachusetts 02451

Received 18 January 2015; revised 12 May 2015; accepted 18 May 2015


Published online 14 July 2015 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.24541

ABSTRACT: Amorphous solid dispersion (ASD) can accelerate a project by improving dissolution rate and solubility, offering dose escalation
flexibility and excipient acceptance for toxicology studies, as well as providing adequate preclinical and clinical exposure. The prerequisite
physicochemical properties for a compound to form a stable ASD are glass-forming ability and low-crystallization tendency, which can
be assessed using computational tools and experimental methods. Polymer excipient screening by in silico miscibility prediction and
experimental screening techniques is discussed. Improved technologies for polymer screening with minimal quantity of drug substance,
and the scalability of ASD from bench to commercial are reviewed. Considerations of in vitro evaluations, preclinical animal selection, and
the translation of the preclinical results to clinical studies are also discussed. Better understanding of how polymers improve the stability
of the amorphous phase in the solid state and how ASD improves bioavailability have facilitated the applications of ASD ranging from
discovery research to preclinical development and further to commercialization. With the understanding of how ASDs are currently used
in the pharmaceutical industry and what challenges remain to be solved, ASD can be applied to solve drug formulation problems at given
research and development stages.  C 2015 Wiley Periodicals, Inc. and the American Pharmacists Association J Pharm Sci 104:3237–3258,

2015
Keywords: amorphous solid dispersion (ASD); bioavailability; dissolution; formulation; polymer; poorly water soluble compound;
solubility; spray drying (SD); stability; supersaturation

INTRODUCTION lipid-based emulsions, self-emulsifying (nano or micro) drug


delivery systems (SNEDDS, SMEDDS, SEDDS), liposomes,
More than three quarters of new chemical entities in phar-
and so on may be viable options, but their utility is lim-
maceutical research are poorly water soluble,1,2 and for those
ited as the drug loading is typically low in these types of
solution formulations are often unachievable. Even when a
systems.
solution can be prepared with high levels of excipients for
In an amorphous solid dispersion (ASD), the solubility of the
pharmacokinetics (PK) studies, it frequently cannot be toler-
drug substance is improved by disrupting its crystalline lattice
ated during efficacy studies using in vivo disease models.3,4
to produce a higher energy amorphous form. Compared with
Complex formulations may produce unacceptable excipient-
other enabling solubilization approaches, an ASD can hold a
related biological effects in toxicology studies.5 Occasionally,
much higher dose for low-solubility active pharmaceutical in-
micronized or nanonized suspensions can solve these prob-
gredients (APIs). ASDs improve bioavailability by maintaining
lems with minimal amounts of excipients. However, they of-
supersaturation in the gastrointestinal (GI) tract. When su-
ten cannot offer adequate exposure because of their slow and
persaturation is sustained, the absorption of a compound can
incomplete dissolution.5,6 When poor water solubility is be-
be maximized to be greater than that of a saturated solution.7
cause of the lipophilicity of a compound, formulations such as
Furthermore, the solubility differences among crystalline phys-
ical forms (e.g., polymorphs) are eliminated because they are
Abbreviations used: API, active pharmaceutical ingredient; ASD, amorphous converted to the amorphous form. This is particularly bene-
solid dispersion; BCS, biopharmaceutics classification system; BMS, Bristol
Myers Squibb company; CAP, cellulose acetate phthalate; CP, coprecipitation;
ficial for compounds in the research stage when the form is
CVD, centrifuge vacuum drying; DDS, dynamic dielectric spectroscopy; DMA, not clearly defined. ASDs are usually well tolerated in ani-
dimethyacetamide; DMF, dimethylformamide; DMSO, dimethyl sulfoxide; DSC, mal disease models and are acceptable to toxicologists as their
differential scanning calorimetry; Eudragit E, L, S, FS, polymethacrylates; FD,
freeze-drying; GI, gastrointestinal; HME, hot-melt extrusion; HPC, hydrox- polymer carriers are derived from GRAS (generally regarded as
ypropyl cellulose; HPLC, high-performance liquid chromatography; HPMC, hy- safe) excipients. Commonly used excipients for forming ASDs
droxypropyl methylcellulose; HPMCAS-L, -M, -H, hydroxypropylmethyl cellulose are cellulose derivatives such as hydroxypropyl methylcellulose
acetate succinate L grade, M grade, and H grade; HPMCP, hydroxypropyl methyl-
cellulose phthalate; HTP, high-throughput; KSD, KinetiSol R
dispersing; MBP, (HPMC), hypromellose acetate succinate (HPMCAS), hydrox-
microprecipitated bulk powder; MiCoS, miniaturized coprecipitation screening; ypropyl methylcellulose phthalate (HPMCP), hydroxy ethyl cel-
NME, new molecular entity; PEG, polyethylene glycols; PK, pharmacokinet-
ics; PLM, polarized light microscopy; PVAP, polyvinyl acetate phthalate; PVP,
lulose, hydroxypropyl cellulose (HPC), cellulose acetate phtha-
polyvinylpyrrolidone; PVP/VA 64, polyvinylpyrrolidone-vinyl acetate copolymer; late (CAP), methyl cellulose, carboxymethyl cellulose, ethyl
RE, rotary evaporation; RH, relative humidity; SC, solvent casting; SCF, spin- cellulose, carboxymethyl ethyl cellulose, chitosan, cyclodex-
coated film; SD, spray drying; SE, solvent evaporation; SEM, scanning electron
microscopy; SLS, sodium lauryl sulfate; ssNMR, solid-state nuclear magnetic trin and derivatives, lactose, poloxamers, polyvinylpyrrolidone
resonance; Tg , glass transition temperature; Tm , melting temperature; UV, (PVP), polyvinylpyrrolidone-vinyl acetate copolymer (PVP/VA
ultraviolet; XRPD, X-ray powder diffraction. 64), polyvinyl acetate phthalate (PVAP), polymethacrylates
Correspondence to: Yan He (Telephone: +781-434-3581; Fax: +781-466-3788;
E-mail: Yan.he2@sanofi.com) (Eudragit E, L, S, FS), and polyethylene glycols (PEG) deriva-
Journal of Pharmaceutical Sciences, Vol. 104, 3237–3258 (2015) tives. These polymers are biologically inert and minimally

C 2015 Wiley Periodicals, Inc. and the American Pharmacists Association absorbed.

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015 3237


3238 REVIEW

Amorphous solid dispersions are generally prepared using should be considered when this system is evaluated in vitro
methods based on solvent evaporation (SE) or melt cooling. and in vivo. A roadmap with the aim to help selection an ASD
The processing technologies include spray drying (SD), hot-melt system is provided.
extrusion (HME), KinetiSol R
dispersing (KSD), drum drying,
freeze-drying (FD), rotary evaporation (RE), spray congealing,
coprecipitation (CP), cogrinding, spin-coated film (SCF), cen- BACKGROUND
trifuge vacuum drying (CVD), supercritical fluid technology, Solid Dispersion
electrostatic spinning, and microwave technology.8–13 HME is
the preferred option in pharmaceutical development because of The first application of a solid dispersion to increase bioavail-
the favorable powder properties generated from this technology, ability was reported over 50 years ago. In 1960, sulfathia-
the absence of organic solvents in processing, small footprint of zole was orally administrated to humans in a eutectic mixture
the equipment, ease of increasing batch size, scalability from with urea.18 The absorption and excretion of the eutectic mix-
pilot to industrial setting, and suitability of continuous process- ture was higher than that of sulfathiazole alone. The authors
ing. However, the application of HME in discovery research is proposed that this new formulation method provided micro-
limited by the amount of API that is required for screening. scopic intimate contact of fine crystallites, which gives an im-
Grams of material are still required to fill the currently avail- proved therapeutic effect of pharmaceutical compounds. They
able laboratory-scale extruders. Even larger amounts of API described a new concept of using a physiologically inactive but
are required for KSD and drum drying. RE is a good choice for easily soluble compound as a carrier to improve the dissolution,
discovery, but it is not a good option when scale-up is needed wettability, and solubility of a poorly soluble active to increase
for projects that progress into development. Supercritical fluid its absorption. After the carrier is dissolved in the intestinal
technology, electrostatic spinning, and microwave technology fluid, the active is suspended as fine dispersed particles, which
are still in their infancy and have yet to be proven in pharma- wet better and have much greater surface area for dissolution.
ceutical R&D. FD can be a good choice when the solvent is wa- The definition of solid dispersion was further refined by Chiou
ter. It is not applicable for scale-up when the API and polymer and Riegelman19 in their review article published in 1971. They
carriers can only be dissolved in organic solvents. In addition, defined solid dispersion as one or more active ingredients in an
the FD process is about 30–50 times more expensive than SD, inert carrier or matrix in the solid state prepared by melting (fu-
which renders FD a cost-ineffective choice for production.14 In sion), SE, or a melting-solvent method. Since then, solid disper-
SD, the use of organic solvents is often unavoidable and this sion has become a platform to overcome the low-bioavailability
raised issues with potential for residual solvents in the ASD, barrier for poorly water-soluble compounds.2,19–24 In this plat-
safety of the workers, and environmental impact of the exhaust form, the dispersion can be a single amorphous phase mixture
gas. Nevertheless, SD is usually the technique of choice to pre- of an active drug with polymer(s), a crystalline mixture of ac-
pare ASD in discovery when drug substance is only available tive with polymer(s), solid complexes of active with complexing
in limited quantities because the screening requires only mil- ligands, or an active dissolved in solid lipid-based excipients.24
ligrams of API. In addition, the development time using SD
Amorphous Solid Dispersion
is also relatively short and a wide range of polymers can be
used. Spray dryers are available not only as bench-top instru- Among the aforementioned solid dispersions, bioavailability
ments that can function using only milligrams of API, but also can be ultimately improved when the active is in an amorphous
as commercial scale equipment that can prepare kilogram to form. Two to three decades ago, researchers led by George Zo-
ton quantities of ASDs. They offer facile scalability from drug grafi and other pharmaceutical scientists, started to exploit the
discovery to development, to commercial production.15–17 solubility advantage of the amorphous forms of drugs in solid
Utilizing spray-dried dispersion in discovery has been a dispersions.25 This type of solid dispersion is specified as ASD.
proven approach to accelerate projects in the pharmaceutical In ASD, the crystalline drug is converted to its amorphous
industry. It opens a new avenue for downstream development, form and stabilized by a polymer carrier. The polymer carrier
yet it does not lock the development to this approach. When not only helps to increase dissolution and solubility of the drug,
more API is available, it is possible for the preparation of ASD to but also to improve the drug’s solid-state physical stability by
be switched to other techniques, such as HME or drum drying. reducing its molecular mobility and increasing its glass tran-
In many cases, ASD is only needed in discovery as an enabling sition temperature (Tg ). It can offer additional benefits when
step. As the project progresses, the drug substance is better the compound is only available as its amorphous form. This
characterized and its solid form is better defined. Switching to strategy has been applied to stabilize the amorphous compound
other formulation approach within the next stage of develop- when it is not chemically stable during storage, shipping, and
ment is often achievable and preferred. manufacture processes.26 It has also been utilized to sustain
The objective of this review is to assess current understand- supersaturation when the neat amorphous compound alone
ing of the chemistry of ASDs, how it is utilized, and what limits cannot.27 The stability of an ASD is likely the result of dis-
its applications. The main focus will be on what physicochem- rupting intermolecular interactions in the drug’s crystal lattice
ical properties of the API determines a candidate’s suitability and forming drug–polymer interactions. Steric hindrance and
for ASD, how the polymer carrier stabilizes the amorphous API hydrophobic interactions can also retard ASD phase separation
in both dry and wet states, how screening is performed to select and API form conversion processes.28,29
the polymer matrix and optimize drug load, how the selection The improved bioavailability resulting from an ASD is be-
criteria for ASDs are applied for projects in the discovery stage lieved to be the results of the synergic effects of thermodynamic
and for later stage products in the pipeline, what should be and kinetic forces. Thermodynamically, there are fewer energy
considered when a project is progressed to the next stage, what barriers as the API is in an amorphous form that is a higher en-
challenges this drug delivery system poses, and what factors ergy state. Its dissolution is more extensive and faster because

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015 DOI 10.1002/jps.24541


REVIEW 3239

the GI tract.

G
Famorphous = e RT × Fcrystalline (1)

where Famorphous is the solubility of the amorphous form,


Fcrystalline is the solubility of the crystalline form, G is the free
energy difference between the amorphous form and the crys-
talline form, R is the molar gas constant, and T is the testing
temperature (K).

Spray Drying
Spray drying is the transformation of a solution feedstock from
a fluid state into a dried particulate form by spraying the feed
into a hot drying medium. The earliest mention of the SD con-
cept was in 1865, and the first detailed description of drying
products in spray form was carried out by Samuel Percy in
a patent granted in 1872.36 The significant utilization of the
SD technology in industries, such as food and detergents, was
Figure 1. Amorphous solid dispersion generates and maintains a su-
not realized until 1920s.37 This technology has been explored
persaturated solution. A: Crystalline drug that has lower solubility.
B: Amorphous drug alone that has higher apparent solubility but pre- by pharmaceutical scientists since 1970s to reduce the particle
cipitates rapidly. C: ASD with weak parachute polymer that cannot size of drug substances and to convert the crystalline form to
delay the precipitation of the active to hold the supersaturation long the amorphous form.38
enough to maximize absorption. D: ASD with strong parachute polymer Compared with fusion methods, such as HME, SD is the
that can sustain the supersaturation for absorption. technique of choice for thermally sensitive compounds as the
drying droplets are at a lower temperature and the drying time
is short.39 Although the temperature for the inlet drying gas
may be as high as 120°C, the temperature of the outlet gas is
it does not need energy to disrupt the crystal lattice. Kinetically,
usually 60°C or lower. In fact, the actual temperature of the
the conversion of the amorphous form back to a crystalline
evaporating droplets is even lower because of the cooling effect
form is impeded by the polymer. The polymer interacts with
caused by the latent heat of vaporization. The drying time is
the molecules of the active, keeping them apart by attractive
extremely short, on the order of milliseconds.
forces (e.g., hydrogen bonding) and steric hindrance. Its large
A large volume of solvent is often required to prepare an ASD
surface area can act as a crystallization inhibitor, delaying or
by SD as the hydrophobic drug, and the hydrophilic carrier has
preventing nucleation and crystal growth. Because of the inter-
to be dissolved in a common volatile solvent. Although finding
molecular contacts of the active with the carrier molecules, the
the common solvent remains challenging, the efficiency of sol-
surface area of the active is also maximized to assist in faster
vent removal in SD is greatly improved over the traditional
dissolution as described by the Noyes–Whitney equation. The
methods, such as RE.40 Compared with a film generated from
wetting is improved because the water-soluble polymer helps
SE and a dispersion prepared by ball milling, the drug and the
facilitate the rapid influx of water into the solid dispersion.
polymer are mixed better at the molecular level in dispersions
In the presence of the polymer, the dissolution kinetics of the
produced by SD.30,41
compound can be greatly improved. Even in the case where
The availability of the instrumentation is an important fac-
complete dissolution and release of the drug from the dosage
tor driving the utilization of a technology. Access to a spray
form is not achieved, the absorption can still be improved when
dryer certainly enables formulation scientists to apply this
a supersaturated solution is generated and maintained during
technology in early research. In the last 20 years, bench-top
its GI tract transit time. ASD can also increase permeation
micro spray dryers, such as Buchi B-90, B-290, B-191, B-190,
rate by the spontaneously formed micro- or nanoparticles or
Niro SDmicro, Anhydro SPX, ProCepT 4M8, and ProCepT 4M8-
micelles in the GI tract.30–32
Trix, have become standard tools in most major pharmaceutical
The mechanism for an ASD to generate and maintain super-
research and development laboratories. The utilization of SD
saturation in the GI tract is illustrated in Figure 1 using the
has been implemented in pharmaceutical companies such as
“spring parachute” analogy, which was introduced by Guzmán
Addex,42 Abbott,43,44 Bristol Myers Squibb company (BMS),45,46
et al.33,34 Depending on the enthalpy and entropy of fusion, the
Merck,5,47 Novartis,48 Pfizer,49,50 Roche,13,51,52 Sanofi,53,54 and
free energy between the amorphous and crystalline forms of a
Vortex.55
compound can be very different. Consequently, the solubility of
the amorphous, as expressed in Eq. 1, can be tens to hundreds
of times greater than that of its crystalline counterpart.35 The
COMPOUND GLASS-FORMING ABILITY AND
extra free energy in the amorphous form behaves like a spring
CRYSTALLIZATION TENDENCY
to bring the concentration of the active to a level that is higher
than that of its crystalline form. The polymer behaves like a Before ASD can be explored as an option to deliver a compound,
parachute to keep the concentration from decreasing. A strong it is important to know whether the compound has suitable
parachute can help hold the transient supersaturated state by physicochemical properties. Researches have suggested that
minimizing precipitation of an active during its residence in the properties of being a good glass former with low tendency to

DOI 10.1002/jps.24541 He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015


3240 REVIEW

recrystallize are the key criteria for determining the potential expansion coefficient when a supercooled liquid is cooled below
for a crystalline drug to be formulated as an ASD.2,8,56,57 approximately 2/3 of its normal freezing point (K). However,
These properties can be experimentally evaluated.57 They the relationship of Tg /Tm = 2/3 was later recognized as the
can also be predicted based on measured or calculated parame- Beaman–Boyer empirical rule, because Beaman and Boyer ap-
ters, such as the melting and glass transition temperatures, plied it to polymer chemistry and their publications led to a
melting enthalpy and entropy, melting viscosity, crystalline greater awareness of this relationship.67,68 The ratio of Tg /Tm
density, molecular size and weight, molecular mobility, flexi- reflects both the thermodynamic and kinetic driving forces. A
bility, complexity, number of benzene rings, level of molecular compound with a high Tm value has strong tendency to crystal-
symmetry, level of branched carbon skeleton, number of rota- lize because of the large thermodynamic driving force, whereas
tional bonds, number of electronegative atoms, and number of a compound with a low Tg value has low kinetic barrier for
hydrogen bonds.58–62 molecular diffusion. This relationship has been widely applied
The amorphous form of a drug substance can often be gen- to polymer materials, alloy materials, and pharmaceutical com-
erated from its crystalline form. The neat amorphous form of pounds with modified versions and refined numbers to predict
a compound can also be obtained from condensation of its va- glass-forming ability.69–72
por, vitrification of its melt mass, or breakage of a crystalline The reciprocal of the Tg /Tm (K/K) value has been applied
form by milling or grinding.63 Once the amorphous property is not only to predicting the glass-forming ability and crystal-
confirmed, its stability can be evaluated under stressed condi- lization tendency of a compound, but also to selecting drug
tions with accelerated temperature and humidity. The forma- loading in ASD.56,71 In the study reported by Friesen et al.,56
tion of crystalline material is commonly monitored with polar- 139 compounds were selected for ASDs because their Tm /Tg
ized light microscopy (PLM), X-ray powder diffraction (XRPD), (K/K) values were less than 1.6 (the reciprocal of this value
or differential scanning calorimetry (DSC). makes Tg /Tm ˜ 2/3). These compounds were divided into three
At the early stage of a project, the access to material is often groups. The first group has the least tendency to crystallize
limited. With a minimum amount of drug substance, a quick with Tm /Tg value less than 1.25. The second group has higher
assessment using modulated DSC with a predefined heating– tendency to crystallize with the Tm /Tg value between 1.25 and
cooling–heating cycle was developed by Baird et al.64 On the 1.4. The third group has even higher tendency with the Tm /Tg
basis of the vitrification and recrystallization behaviors in the value between 1.4 and 1.6. Using HPMCAS as the polymer
cycle, 51 randomly selected organic molecules were classified carrier, the first group compounds were successfully formu-
into three classes, as summarized in Table 1. The cooling and lated as ASD with 50% drug load, the second group compounds
reheating rates are critical in this screening, because they gov- were formulated with 35%–50% drug load, and the third group
ern vitrification, formation of nuclei, and crystalline growth. compounds were formulated with 10%–35% drug loading. The
Crystallization tendencies between molecules of similar struc- success of developing ASD for MK-0364 using hydroxypropyl
ture were found to vary significantly. The understanding of methylcellulose acetate succinate L grade (HPMCAS-L) was
the driving force for crystallization in each class of compounds also attributed to the good glass formation ability of MK-
was further investigated by Kawakami et al.65 with a dataset 0364, which has Tg 41°C, Tm 104°C, and Tm /Tg (K/K) 1.20.47
of seven compounds. For Class I compounds, the crystallization Though the physical stability with the defined drug load can
feature was determined to be dominated by thermodynamic fac- be predicted for ASD using the relationship of Tm /Tg , the ef-
tors (temperature). The crystallization of Class II compounds fect of drug load on in vivo absorption needs to be studied and
was found to be influenced by competition between thermo- confirmed.
dynamic and kinetic factors. Although the crystallization of Using a single parameter to predict ASD stability may over-
compounds in this group is still dominated by temperature, simplify the reality. However, this ratio can serve as a quickly
the transformation is perturbed by steric hindrance that can accessible starting point to understand whether a compound
be measured as local pressure. There is a large energy barrier may be a suitable candidate for ASD, as Tm s and Tg s for known
for compounds in Class III to nucleate, which leads to a low compounds are abundant in the literature and can be easily
probability for them to crystallize. The dominant factor for the measured for new compounds using a modulated DSC with a
crystallization of compounds in this group is pressure. It was small amount of API.
also concluded that for the compounds in the high-crystalline In the absence of experimental data input, sophisticated
tendency groups, Class I and II, the initiation time for their computational tools can also provide initial insights. An al-
crystallization could be generalized as a function of the ratio of gorithm solely based on calculated molecular descriptors was
Tg to T (the storage temperature), Tg /T. In a recent publica- developed by Mahlin et al.59 to predict glass-forming ability. The
tion, Trasi et al.62 investigated glass stability and concluded input for this model was 245 variables obtained from DragonX
that the following physicochemical properties can contribute (Talete, Italy) using partial least squares projection to latent
to fast crystal growth rates: low molecular weight, high melt- structure discriminant analysis. The response variables were
ing temperature (Tm ), low melt entropy, low melt viscosity, and glass former (assigned value 0) and nonglass former (assigned
high crystal density. value 1). This model suggested that larger molecules with a low
The Tm , Tg , and their ratio have been used to predict glass- number of benzene rings, low level of molecular symmetry, and
forming ability and crystallization tendency, as Tm is a well- branched carbon skeleton and electronegative atoms have the
accepted parameter to evaluate the thermodynamic driving ability to form a glass. The glass formation prediction was ex-
force for crystallization, and Tg is a popular measure to as- perimentally verified on a training set of 16 compounds whose
sess the kinetic barriers of the amorphous. The relationship amorphous forms were generated using SD, melt quenching, or
of Tg and Tm was first identified by Kauzmann.66 He pointed mechanical activation. Only one compound failed in the predic-
out the 2/3 rule, which he stated as a glass transformation tion. When a random selected testing set of 16 compounds was
event with sudden drop in the specific heat and the thermal checked, there was a 75% success rate.

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015 DOI 10.1002/jps.24541


REVIEW 3241

Table 1. Classification of Glass-Forming Ability and Crystallization Tendency by DSC64,65

Group Cooling Reheating Classification Crystallization Driving Force

Class I Crystalline – Poor glass former/high crystalline tendency Thermodynamic


Class II Amorphous Crystalline Good glass former/high crystalline tendency Thermodynamic and kinetic
Class III Amorphous Amorphous Good glass former/low crystalline tendency Pressure

A follow-up study by this group of researchers extended to predict the thermodynamic miscibility between drug and
the prediction with rapidly measured physical properties from polymer.73,80–84
DSC.60 The authors suggested that a decision making on the
selection of amorphous or ASD formulation can be rationalized G Ndrug Npolymer
based on the molecular weight, and the predicted or experimen- = ln Ndrug + lnNpolymer + PNdrug Npolymer (2)
RT mdrug mpolymer
tally obtained glass transition and recrystallization tempera-
tures. Although low crystallization tendency in the dry state is
recognized as an important factor for the ASD shelf life, low where G, R, and T are the same as in Eq. 1, Ndrug is the volume
crystallization tendency in the wet state is crucial to achieve fraction of drug, mdrug is the ratio of the volume of the drug to
improved bioavailability.57 the lattice site, Npolymer is the volume fraction of polymer, mpolymer
is the ratio of the volume of the polymer to the lattice site, and
P is the Flory–Huggins interaction parameter representing the
POLYMER SCREENING FOR SOLID DISPERSION difference between the drug–polymer hetero contact interac-
tion, the drug–drug homo contact, and the polymer–polymer
Even for compounds that are identified as good glass form-
homo contact interactions.
ers with low crystallization tendencies, their amorphous forms
This equation is particularly useful in describing drug–
are still thermodynamically unstable. Incorporating the amor-
polymer mixing systems because it takes into account the
phous form into a polymeric solid dispersion can drastically al-
large size discrepancy between the drug molecule that usu-
ter the kinetics of conversion to a crystalline form in both solid
ally has molecular weight less than 600 Da and the polymer
and supersaturated solution states. The physicochemical prop-
molecule that usually has molecular weight between 10,000
erties of the drug and the polymer determine how the polymer
and 1,500,000 Da. The first two terms in the right side of the
interacts with the drug molecule in the ASD. The ideal polymer
equation represent entropy, which always favors mixing. The
plays multiple roles in the dispersion. First, it has the proper-
magnitude of the entropy upon mixing is relatively constant
ties to maintain the drug’s amorphous state not only during the
in such systems where small size molecules mix with much
manufacturing but also during shipping and storage. Second,
larger molecules. The miscibility of the drug–polymer system is
it dissolves rapidly and releases the drug at the absorption site
therefore determined by the Flory–Huggins interaction. When
in the GI tract. It also has the ability to maintain the super-
the drug–polymer hetero contact interaction is greater than the
saturated solution for an adequate period to allow absorption.
summation of the drug–drug and polymer–polymer homo con-
Furthermore, it may also enhance the permeation of the com-
tact interactions, the value of P will be negative, which means
pound through the GI tract membranes.
the system also enthalpically favors mixing, and hence the
The miscibility of the drug and polymer is generally believed
drug–polymer system is miscible. In the case that the drug
to be the prerequisite condition to form a physically stable ASD.
and the polymer lack significant intermolecular interactions,
A miscible dispersion is defined as one single amorphous phase
enthalpy does not favor mixing. However, the system can still
system, which has one single Tg and no detectable phase sepa-
be miscible if the mixing entropy is sufficient to offset the un-
ration by analytical techniques, such as DSC, XRPD, infrared
favorable adhesive intermolecular interactions.
spectroscopy, solid-state nuclear magnetic resonance (ssNMR),
This theory serves as a good starting point to understand the
scanning electron microscopy (SEM), dynamic dielectric spec-
thermodynamics between drug and polymer.80 Although it is a
troscopy (DDS), and Raman.24,73–77 A great deal of research
useful theory for the prediction of drug solubility in polymer
efforts have been employed in predicting the compatibility and
carrier, its limitations are also well-known because this theory
the phase stability of drug–polymer mixtures. These studies
is based on mean-field approximation to facilitate the calcu-
are shedding light on the stabilization mechanisms of the amor-
lation for placement of a polymer molecule in a partly filled
phous compound in an ASD. However, because of the complexity
lattice, the assumptions to generate this equation are based on
of the interactions, deviations of experimental results from the-
polymer–polymer or polymer–solvent systems, and the energy
oretical predictions are still ubiquitous. In industry, the selec-
for breaking the crystalline lattice is not included.78,81
tion of the optimal polymer still mainly relies on experimental
Solely based on the chemical structures of the drug and the
screening.2
polymer, the value of P can be calculated from solubility param-
eters as in Eq. 3.79,85,86 The solubility parameters for the drug
Theoretical Drug–Polymer Miscibility Prediction
and the polymer can be estimated from a group contribution
It is recognized that there is not a well-established method method.81,82,84,87 This method has been incorporated into some
available to measure the miscibility between drug and poly- commercial prediction tools, such as the MemFis R
software, to
mer because the polymer’s viscosity impedes the attainments allow in silico polymer screening. The accuracy of these in silico
of equilibrium. Currently, it can only be estimated by model predictions need to be experimentally verified as the lack of pre-
prediction.78 The lattice-based Flory–Huggins theory,79 ex- dictive power is known when the solubility parameter is used
pressed in Eq. 2, has been adapted from polymer blends to predict systems where the interactions are predominated by

DOI 10.1002/jps.24541 He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015


3242 REVIEW

forces other than van der Waals forces. Highly directional in-
teractions (e.g., hydrogen bonding) and long-range interactions
(e.g., ionic interactions) are known to form in drug–polymer
systems.88

L(*drug − *polymer )2
P= (3)
RT

where R, T, and P are the same as in Eqs. 1 and 2, L is the


volume per lattice site, and * is the solubility parameter for the
drug, *drug , and for the polymer, *polymer .
When the calculation was coupled with experimental data,
much improved predictions can be achieved. The experimental
data can be the melting point depression in the mixture of
the drug and the polymer or solubility of the drug in a lower
molecular weight analog of the polymer.73,80,81,83

Experimental Screening
Although there are improved techniques for the prediction of
the solid-state stability of ASD, the prediction of ASD disso-
lution performance remains challenging because it remains
difficult to predict the ability of a polymer to prevent drug crys-
tallization in supersaturated solution. At present, the identifi-
cation of polymeric carriers that increase dissolution rate and
also stabilize supersaturation remains largely empirical.2 Ex-
perimental screening of a polymer is typically unavoidable.

Solvent Shift Figure 2. Flow diagram of HTP screening for polymer and drug load-
ing by solvent casting and dissolution method.
The solvent shift technique, also known as cosolvent or solvent
quench method, is traditionally used to evaluate the ability
of a polymer to attain supersaturation and to prevent precip- optimum drug load will be. Even in the case where the polymer
itation from supersaturated solution.29,89–91 The polymers of can form an ASD with the compound, the dissolution behavior
interest are dissolved in aqueous media, which can be wa- of the ASD can diverge sharply from the screening results. For
ter, buffer, or simulated biorelevant fluid. The drug is dis- example, in one solvent shift study, the AUC of itraconazole in
solved in a water-miscible organic solvent, such as ethanol, a buffer solution with hydroxypropyl methylcellulose acetate
propylene glycol, PEG, dimethylformamide (DMF), dimethyac- succinate H grade (HPMCAS-H) was about seven times that
etamide (DMA), N-methylpyrrolidone, 1,4-dioxane, or dimethyl in buffers with HPMCAS-M or HPMCAS-L. However, the dis-
sulfoxide (DMSO). In addition to organic solvent, an acidic drug solution AUC from the ASD that was made with HPMCAS-H
can be dissolved in alkaline media, such as sodium hydroxide was only about 2% of that from the ASDs that were made with
solution, and a basic drug can be dissolved in acidic media, such HPMCAS-M or HPMCAS-L. Although the rank order to stabi-
a hydrochloric acid solution. The drug solution is then added lize itraconazole supersaturated solution from the solvent shift
dropwise into the aqueous media containing the test polymer. method was H > M ≈ L, the rank order to promote dissolution
The target can be a predetermined drug concentration or until from ASD was L ≈ M  H.29
precipitation is noticed. The final solution contains a minimal
Solvent Evaporation and Casting
amount of organic solvent, and the drug concentration should
be greater than its solubility in the media. The precipitation When a drug ASD needs to be produced to assess its dissolution
(appearance of turbidity) can be monitored visually or detected behavior, a rotary evaporator becomes a useful tool as it is
via nephelometry or by a focus beam reflectance measurement. available in almost every research laboratory.92–95 As a thin
The concentration of drug in solution can be monitored by an film is formed after the SE, this method can be referred to
in situ fiber optical probe, or measured by ultraviolet (UV) or as solvent casting (SC). A film can also be cast on a Teflon-
high-performance liquid chromatography (HPLC) using super- coated glass plate or a silicon chip after rapid spinning.41,96
natant that is withdrawn at predetermined time points and This screening can be performed in a HTP manner.51,97,98
then filtered or centrifuged. As described in the Solvent Evap- The HTP SC process is schematically depicted in Figure 2.
oration and Casting section, this screening can be performed The test compound is dissolved in a suitable organic solvent
in a high-throughput (HTP) fashion with robotic automation as stock solution and dispersed into each well of a 96-well
using multiwell plates. The best-performing polymer is the one plate. The contents can be the same compound for the entire
that offers the highest drug concentration in solution for the plate with the same or different volume for the same or differ-
longest time. The results help to select the polymer that stabi- ent drug load. The contents can also be multiple compounds
lizes the compound’s supersaturation. These screening results for one plate leading to fewer combinations. The test poly-
alone, however, do not provide information on whether the poly- mers are also each individually dissolved in a suitable organic
mer is capable of forming an ASD with the drug or what the solvent and dispensed into these wells. A second polymer or

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015 DOI 10.1002/jps.24541


REVIEW 3243

surfactant can be added if the scope includes ternary or higher- manner into 1 mL glass vials, which are inserted in the wells of
order combinations. After dispensing, the plate is covered and a 96-well plate, as in Figure 2. The vials are filled with acidified
mixed via vortexing or a method designed for the robotic work- water (e.g., 0.01 N HCl), chilled at 5°C, and stirred to produce
station. Then the solvent is removed, most commonly via vac- fine microprecipitated bulk powder (MBP). The suspensions
uum evaporation. A solid film is expected to form at the bot- are next transferred to filter plates, washed, dried, and then
tom of the well. The plate can be characterized by techniques follow the same steps as in Figure 2, where the powder can be
such as PLM, SEM, XRPD, or Raman. As described by Chiang characterized, dissolved, and analyzed.101
et al.,51 the plates can be prepared as duplicates with one for The MBP preparation was adapted from a general CP
characterization and stability studies while the other is used method by using enteric polymers.13 This method is suitable
for solubility studies. When only one plate is prepared, the plate for drugs that are not amendable to HME, SD, RE, or FD be-
can first be characterized for amorphous contents, then evalu- cause of high Tm , thermal instability, or low solubility in volatile
ated for stability for a given time frame, characterized again, organic solvent and water. Although the laboratory-scale MBP
and then used for dissolution testing. The goal of the screen- experiment requires at least 100 mg API to test one condition,
ing is to select a polymer that can form solid dispersion with this integrated miniaturized system can be used to screen ASDs
the testing compound to improve its dissolution. When a buffer with as little as 2–10 mg API per sample.101
is added to the plate, the solid film will be hydrated, and the
dissolution of the compound can be facilitated with shaking or Single Droplet Drying
stirring. At a predefined time point, usually 1–3 h, the contents
Some research laboratories are working with the single droplet
are transferred to filter plates and the filtrates are diluted with
drying technology to produce amorphous forms with even lower
an organic solvent to prevent further precipitation, and finally
drug substance requirements for screening.
the concentrations are measured by UV or HPLC.
Aerodynamic, electromagnetic, and acoustic levitation have
The screening can be modified along the process line at any
been explored to develop amorphous drug on single droplet
step to accommodate the needs of the project.51,97–99 This HTP
individually dried particles.102,103 Drug solutions that have a
screening method has been adapted by many contract research
propensity for forming viscous fluids have been screened on an
and manufacturing organizations, such as Evonik and Bend
acoustic levitator by SE or laser heating.104 During levitation,
Research, to screen polymers for customers. In the work of Bar-
droplets with 1–3 mm diameters are introduced and dried while
illaro et al.,97 HTP was performed on binary combinations of
floating freely in the gas phase. The drying process is similar
phenytoin with polymer or surfactant at drug loadings of 10%,
to the SD process as the droplets do not contact any surface.
20%, or 40%. Three polymers at all three tested drug loads
Therefore, the extrinsic heterogeneous nucleation is avoided
that provided dissolution greater than 90% after 30 min were
and the results are expected to be reproduced in scaling up us-
selected for scale-up using the RE method. The dissolution pro-
ing SD.103 The individually dried droplets of griseofulvin with
files of the RE scaled-up ASDs correlated well with those ob-
2.5% and 20% drug load in PEG 6000 mixtures were levitated
tained from HTP screening.97
in a drying kinetics analyzerTM .105 Their physical and chemical
Chiang et al.51 reported that SD was used to successfully
properties were compared with bulk spray-dried powders com-
scale up an ASD based on HTP screening results. In their
posed of the same ratios of the griseofulvin and PEG 6000. The
study, HTP screening was performed on four model compounds
characterizations were performed by atomic force microscopy
with loadings of 10%–70% using HPMC K100, HPMCAS-M,
imaging and confocal Raman microscopy. For the same drug
or PVP K90 as the carrier. Duplicate plates were prepared.
load, it was found that the individually dried particles were
Full characterization was performed on the plates that were
equivalent to the bulk spray-dried powders. Using this tech-
stored at 50°C/75% RH (relative humidity) for 2 weeks. The
nology, with very minimal amount of material, drug–polymer
HTP screening method was validated by a low-solubility b-Raf
combinations can be screened to simulate SD. However, only
kinase inhibitor, Compound A. The physical stability and sol-
a few pharmaceutical laboratories are currently equipped with
ubility after 2 weeks stored at 50°C/75% RH were evaluated.
such instrumentation. Literature on the use of the single par-
The data generated by the HTP method were in good agreement
ticle (or droplets) to screen formulations is limited.
with that generated from the spray-dried material. HPMCAS-
M with 20% drug load offered the highest solubility improve-
Spray Drying
ment. In a separate publication, the spray-dried Compound
A with 25% drug load in HPMCAS-M offered much improved Although both SD and SC produce materials via SE, the films
bioavailability.100 The authors attributed the success of scaling and the spray-dried particles can produce very different dis-
up the HTP method to SD by the rapid evaporation in their solution profiles.47,105 Even in the case where the miscibil-
HTP experimental conditions. A high-vacuum system and heat ity limit is exceeded, rapid drying process still most likely
were utilized to ensure rapid solvent removal. Using the Hick- generates well-mixed drug–polymer systems in spray-dried
man and Clausius-Clapeyron equations, they concluded that it products.78 With micro spray dryers becoming increasingly
only takes 1–15 s to completely evaporate the organic solvents common in pharmaceutical laboratories, many pharmaceuti-
in these wells. cal scientists choose to directly screen formulations using SD
technology.5,42,45,106
The B-90 is the fourth generation of laboratory-scale spray
Miniaturized Coprecipitation Screening
dryer from BL̈chi Labortechinik AG. It offers 40%–95% yield
In this screening technique, the API and polymer stock solu- for 30–500 mg of material.107–110 Therefore, 30 mg of compound
tions are prepared in a common water miscible solvent, such with 20% drug load in polymer can be screened using Buchi
as DMA, DMSO, or DMF. They are mixed at predefined drug B-90 to generate about 75 mg product with the assumption of
loading ratios. The mixtures are then dispersed in a dropwise 50% yield. The Bend Mini Spray Dryer from Bend Research

DOI 10.1002/jps.24541 He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015


3244 REVIEW

Inc. is able to provide 82% yield on 50 mg batch size, with 10% consistency. The simulation parameters should include compo-
and 25% drug load, using 5 mL solvent.27 The ProCepT 4M8- sition of the medium, pH of the medium, dose to GI fluid volume
TriX states a greater than 90% yield from as little as 1 mL ratio (with the introduction of the whole intended dosage form
of solution. The introduction of ultrasonic nozzles to bench-top in the dissolution medium), and the exposure (experimental)
spray dryers has improved their capabilities for formulation time. The pH of the medium at the end of experiment should be
screening. Compared with the two-fluidic nozzle configuration, measured. The precipitate, if there is any, should be character-
an ultrasonic nozzle offers the advantage of spraying very small ized. Meanwhile, nonsink condition should be used, although
amounts of solution with better yield by avoiding solution ad- using sink condition is common practice when dissolution is
hesion to the chamber of the small laboratory dryer. Equipped performed for quality control of conventional formulations.114
with ultrasonic nozzles, bench-top spray dryers produce par- The commentary authored by Newman et al.24 thoroughly ana-
ticles with the size, morphology, and powder properties that lyzed publications up to 2010, which reported bioavailabilities
closely resemble those from large spray dryers. This adaption using ASDs. Their analysis and discussion focused on 40 stud-
certainly helps streamline scale up from research to develop- ies, where the drug is in an amorphous form and the polymer
ment and further to product commercialization.16 is the main component of the dispersion (interested readers
As an example, solid dispersions of amlodipine using dex- are urged to consult the article for details). The authors ana-
trin as the matrix with or without the permeability enhancer lyzed all of the reported dissolution conditions and found that
sodium lauryl sulfate (SLS) were manufactured by using a 71% of them used Apparatus II vessels, 92% used nonspecific
Buchi 190 mini spray dryer. The best-performing dispersion or nonsink condition, 90% performed in 500–900 mL media,
demonstrated a comparable rat PK profile to the marketed 88% performed at 50–100 rpm, 50% used media with pH close
product Norvasc R
, which is formulated using amlodipine be- to neutral, 79% used buffer or water, and 21% used simulated
sylate salt.106 In another example, an ASD containing a GPR- gastric fluid or simulated intestinal fluid without enzymes. The
119 agonist was selected as a preclinical candidate formulation, dissolution media were widely varied in composition with pH
which was prepared by a SD process using the ProCepT micro from 1.2 to 7.2.
spray dryer. The formulation selection was based on solid-state In one study, felodipine ASDs were prepared with HPMC as
stability of the ASD upon storage, physical stability of the ASD the carrier.113 The controlling factor in their dissolution was
suspension in dosing vehicle, dissolution in biorelevant media, found to change from the physicochemical properties of the
rat PK profile, and the acceptance of the excipients for preclin- polymer to the solubility of the amorphous drug layer when
ical toxicity studies.5 the drug load in the ASD was increased from 10% to 50%.
The generality of the conclusion was verified by studies of the
same percentage drug-loaded ASDs with either felodipine or
PERFORMANCE OF SOLID DISPERSION indomethacin as the drug and either HPMC or PVP as the
Once solid dispersion is identified as the enabling drug delivery carrier. Using a UV dip probe detector, the peak concentra-
tool for a drug candidate and a polymer or polymer mixture is tions in the dissolution profiles of the ASDs with 10% drug
selected through screening, the solid dispersion containing the load showed full release from the introduced solids. The study
compound can be prepared by a selected processing technique. was performed using various amounts of ASD solids that con-
SD and HME are the most commonly used processes in re- tained the equivalent felodipine concentration of 30, 60, and
search and development, respectively. The solid-state stability 90 :g/mL. However, the dissolutions of the ASDs with 50%
can be evaluated under stressed conditions with accelerated drug load were much less with only about 6% release from the
temperature and elevated RH. The performance of the drug introduced solids. The felodipine concentrations in the disso-
dispersion is determined by the rate and extent of drug release lution profiles of 50% drug-loaded ASDs were close to its theo-
as measured by in vitro dissolution and confirmed by in vivo retically calculated solubility of the neat amorphous drug form.
exposure. Within a few minutes after the 10% drug load ASDs were intro-
duced into the dissolution media, the solids disappeared and the
In Vitro Evaluation media became turbid. In contrast, the media and the solids re-
mained unchanged throughout the experiment (observed under
Dissolution
cross-polarized microscopy) after the 50% drug load ASDs were
Dissolution is the most widely accepted tool to predict in vivo introduced. Dynamic light scattering analysis of the turbid me-
biological performance of a formulation.24 It remains challeng- dia revealed the formation of nanoparticles. The UV spectra
ing to correlate the in vitro dissolution results to the in vivo analysis of these particles indicated that they were crystalline
absorption especially for formulations that produce supersat- nanoparticles of felodipine, which were precipitated from the
uration, because the in vitro dissolution may not be predic- supersaturated solutions. In order to verify the degree of su-
tive of precipitation or fully demonstrate the driving force for persaturation in the dissolution of the ASD, a diffusion study
absorption generated by the solubilizing power of such drug using a dialysis membrane was performed on the dissolution
delivery systems.111,112 This is especially true for ASDs, be- of these ASDs, along with artificially supersaturated felodip-
cause complex processes take place simultaneously during their ine solutions, dissolution of felodipine neat amorphous form in
dissolutions.113 the presence of polymer in the medium, and a saturated so-
The dissolution method should be developed to fit the pur- lution of crystalline felodipine. The equilibrium solubility of
pose at the stage of the development. As pointed out by New- the felodipine crystalline form is about 1 :g/mL, which was
man et al.,24 during the discovery and early phases of drug used as the reference. The artificial supersaturated solutions
development, the dissolution conditions need to be carefully were added at concentrations of 5 and 10 :g/mL in the donor
designed to closely simulate the GI tract environment in order chambers and their resultant measured relative fluxes were 4.8
to predict in vivo performance, not to control batch to batch and 10, respectively. The measured values and their ratios to

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015 DOI 10.1002/jps.24541


REVIEW 3245

the reference indicated that the experimental design accurately In Vivo Evaluation
discriminated between solution concentrations. The measured
Preclinical
solubility of the neat amorphous form in the donor chamber
was about 10 :g/mL and its solution activity calculated from The selection of a preclinical animal model is critical because
the flux value across the dialysis membrane was about nine significant variations in drug absorption can be caused by
times to that of crystalline form. These results were remark- differences in aspects of anatomical, physiological, luminal
ably close to the theoretically calculated values. In contrast to fluid contents, gastric emptying, metabolic, biochemical, mi-
approximately 2.5 times difference in the apparent concentra- crobiome compositions, and so on.115 In the aforementioned
tions in the donor chambers, the flux of the ASDs (11 and 10 commentary,24 four animal species were found in the studies
for the 10% drug load and 50% drug load, respectively) were analyzed: dog (50%), rat (30%), rabbit (18%), and monkey (2%).
similar. These results indicate that there is no significant dif- In literature published after 2010, rat appears as a more pop-
ference in the thermodynamic activity of felodipine in solutions ular model.27,32,42,97,116,117 Other species found in ASD in vivo
derived from the 10%, 50% drug load ASDs or the neat amor- evaluations are mouse, minipig, and pig.24,118
phous API. A solution NMR study on the dissolutions of these In order to generate data that can help predict human expo-
ASDs confirmed the diffusion results. Although the finding in sure and enable human dosage estimation, a clinically relevant
this research is intriguing, a further investigation on in vivo animal model should be selected with clearly defined selection
exposure of these ASDs would be very helpful to shed light on of gender, fed or fasting state, and so on. If fasted, the experi-
understanding how the nanoparticles in the low drug-loaded menter needs to define the fasting time prior to dosing and after
ASD, and the unchanged amorphous compound in the high dosing. If fed, the experimenter needs to control the diet. In the
drug-loaded ASD affect the bioavailability. survey performed by Newman et al.,24 80% of the bioavailabil-
ity studies were performed under a fasted state, whereas free
Permeation food access was allowed for 7% studies and 9% studies were
performed under a defined fed state.
The permeation rate of ASDs has been evaluated by some re-
searchers because they believe this delivery system may also Clinical
alter the permeation rate of a drug. The intracellular uptake The ultimate goal of developing an ASD formulation is to obtain
of 9-nitrocamptothecin was significantly increased on Caco-2 satisfactory exposure in human clinical trials and eventually
cells when its freeze-dried ASD with a drug load of 6% in Solu- patients.
plus was studied. The increased permeation rate was attributed There is no doubt that in vitro and in vivo evaluations are
to the spontaneously formed homogenous nanosized micellar necessary for formulation selection in order to advance the most
structures.32 One study on an ASD prepared by HME contain- appropriate formulation for clinical trials. Both experimental
ing 5% of the model compound ABT-102 in polymer PVP/VA 64 design and data analysis require care. When itraconazole ASD
and surfactants Tween 80, pluronic 188, and sucrose palmitate, with 40% drug load was evaluated in dissolution using the com-
led to a controversial discovery. After dissolution of the ASD, it mercial product Sporanox R
as the reference, it released much
was found the medium contained free API, micelles, polymer- slower from HPMC-based ASD compared with Eudragit E100
bound and solubilized API, as well as microparticulate struc- or a mixture of Eudragit E100 and PVPVA64 based ASD. How-
tures that were mainly formed by amorphous API. Although ever, in a clinical study, the best bioavailability measured by
the increased apparent solubility was attributed to the micel- AUC and Cmax was obtained from the HPMC-based ASD.119
lization and complexation of solubilized API, the increased per-
meation rate was accredited to the amorphous API micropar-
ticulates, which was described as true supersaturation.31 DISCUSSIONS
Some researchers have adapted ASD to improve bioavail- Achievements
ability of biopharmaceutics classification system (BCS) IV com-
pounds by incorporating absorption enhancers into the disper- As demonstrated in this review, academic researchers and
sion. Using orthogonal design, nine combinations of ternary industrial pharmaceutical scientists have shaped our under-
ASDs were prepared for berberine with one of the three poly- standing of the fundamental mechanisms of how polymers in-
mers, PVP K30, PEG 6000, or F 68 as the carrier, and sodium hibit and/or retard the amorphous to crystalline phase change
caprate as the absorption enhancer.95 The membrane perme- in both the solid state and in supersaturated solutions. Table 2
ability of these preparations was assessed using Caco-2 cells, lists some examples that have been discussed in this paper.
and the intestinal absorption was examined in situ using rat
ASD Solid-State Physical Stability
small intestine. The in vivo bioavailability and bioactivity stud-
ies confirmed that the preparation that offered the best perme- Stabilization of an amorphous drug in an ASD is extremely
ation rate and absorption in the in vitro and in situ studies important because the dissolution and exposure would not be
had markedly increased bioavailability and significantly im- improved if the drug reverted to its crystalline form. Even in the
proved antidiabetic effect. SLS at the concentration of 0.9% was case where the drug and the polymer are miscible, their ther-
added to spray-dried amlodipine–dextrin solid dispersion.106 modynamic equilibrium is rarely reached during ASD prepara-
The amount of this tertiary component did not affect the dis- tion. The solid is arrested in a metastable state regardless of
solution of amlodipine. However, the permeation rate of the whether it is rapidly cooled from the melt, rapidly evaporated
ternary ASD was increased on Caco-2 study. Consequently, the from solution, or obtained from other techniques. In addition,
exposure of this ternary ASD in rats was increased compared as long as a supersaturated single phase dispersion is achieved,
with the neat amlodipine powder, and the binary amlodipine– pharmaceutical ASDs are often prepared with drug loads in ex-
dextrin ASD. cess of their equilibrium solubilities in the polymer to satisfy

DOI 10.1002/jps.24541 He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015


3246

Table 2. Examples of Polymer Selection and ASD Evaluation


REVIEW

Compound Structure Polymers (Drug Load, %) Method Selection Criteria Performance Reference

Amlodipine, MW 408.9, Dextrin, dextrin with SD Dissolution, membrane Permeability was better from the 106
Tm 144°C, log P 3.0, 0.9% SLS (10%) permeability, rat PK ternary ASD. Rat PK was also
pKa 9.0 better and comparable to market
product.
Berberine, MW 336.4, Tm PVP K30, PEG6000, F68 RE Solubility, dissolution, All ASDs increased solubility and 95
145°C, log P 0.05, with 3%–33% sodium membrane dissolution while 1:1:6
neutral caprate (13%–44%) permeability, in situ berberine–sodium
intestinal perfusion, caprate–PEG6000 performed the
rat PK best. It provided 3×, 4×, and 5×
increase in permeability, perfusion,
and rat oral bioavailability. No rank
order of polymer can be concluded
because of the experimental design.
BMS-A, MW 500, Tm NA PVPVA, HPMCAS-M Solvent shift, Solubility, dissolution, More solubility was increased by 112
160°C, Tg 45°C, (40%) SD dog PK HPMCAS-M in solvent shift. In

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015


neutral both nonsink and sink dissolutions,
PVPVA ASD performed better.
Better dog PK was obtained from
HPMCAS-M ASD.
BMS-B, MW 688.2, Tm HPMCAS-M (10%, 25%) SD Dissolution (10% and ASD with 10% and 25% drug load 27
amorphous, Tg 88°C, 25% drug load), rat dissolved 90% drug in 7 min and
log P 7.7, pKa 7.6, 9.7 and monkey PK and 25 min, respectively. Satisfactory
TK (25% drug load) and dose-proportional exposure was
obtained in rat and monkey
toxicology studies.
Compound 1, MW 377.5, HPMCAS-L, HPMCP SD Dissolution, rat PK HPMCAS-L ASD crystallized in the 5
Tm 168,173°C, Tg 36°C, HP55 (35%) vehicles of suspension formulation.
log P 3.7, pKa 1.9, 3.3 HPMCP ASD increased 7-fold AUC
from crystalline suspension in
dissolution and exposure in rat PK.
Compound A, MW 425.4, HPMC K100, PVP K90, HTP SC, SD Solubility (HTP Solubility was increased the most by 51,100
log P 1.8, pKa 2.2, pKa HPMCAS-M, materials), rat PK (SD HPMCAS-M, followed by HPMC
8.5 (acidic) (20%–70% in HTP) material) K100, then PVP K90. Solubility was
(25% in SD) the highest at 20% drug load and is
reduced with increasing loading in
all cases. ASD with 25% drug load
provided satisfied rat PK.

Continued

DOI 10.1002/jps.24541
Table 2. Continued

Compound Structure Polymers (Drug Load, %) Method Selection Criteria Performance Reference

DOI 10.1002/jps.24541
Felodipine, MW 384.3, Tm PVP K29/K32, HPMC, SCF Crystallization tendency, These polymers equally effectively 28
147°C, Tg 45°C, log P HPMCAS-M reduced nucleation rate in the
4.8, pKa 2.7 (75%–97%), absence of moisture.
Same as above, (75%) SCF, RE Nucleation rate Equally effectively reduced nucleation 92
rate at 75% RH, PVP absorbs more
water and caused the tendency of
phase separation.
Same as above, RE Dissolution HPMCAS-M produced the highest 93
(2%5–90%) extent of supersaturation, followed
by HPMC, then PVP.
PVP K30, HPLCAS-L SD, HME, Dissolution HPMCAS-L produced higher drug 120
(25%, 33%, 50%), released rates and better
wettability. In both polymer
matrixes, SD materials were
released faster.
HPMC, PVP (10%, 50%), RE, Dissolution, diffusion, Although there is a higher apparent 113
solution NMR dissolution from the 10% drug load
ASDs, a similar maximum
thermodynamic activity of the
dissolved drug was detected from all
ASDs.
Eudragit L100 and MiCoS XRPD characterization All polymers at all drug loads formed 101
L100–55, HPMCAS-L, ASDs.
(20%, 35%, 50%)
Glyburide, MW 494.0, Tm HPMCP HP-55, Eudragit MiCoS XRPD characterization, Only L100 at drug load 40% and 50% 101
169°C–170°C, Tg 65°C, L100 and L100-55, solid stability, did not form ASDs. Although
log P 3.1, pKa 13.7 HPMCAS-L (10%, dissolution L100-55 ASDs offered the best solid
20%, 30%, 40%, 50%) stabilities, HPMCAS-L ASDs
offered the best dissolutions.
Griseofulvin, MW 354.8, HPMCAS-M (20%) FD, SD, CVD Effect of manufacture They offered comparable dissolution 121
Tm 216°C, Tg 88°C, process, rat PK profiles and similarly improved rat
log P 3.0, neutral PK.
Itraconazole, MW 705.6, HPMC E5 and E50, PVP HME Dissolution, rat PK, Among pH-independent polymers, 122
Tm 166°C, Tg 50°C, K12 and K90, Eudragit HPMC was a better stabilizer than
log P 5.7, pKa 3.7 L 100-55, HPMCP 55 PVP, while increasing molecular
and 55S (33%) weight promotes supersaturation.
Among enteric-coating polymers,
Eudragit was better than HPMCP.
Only HPMC E50 and EUdragit L
100-55 ASD were evaluated in rats;
they both performed well.
REVIEW

Continued

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015


3247
3248
REVIEW

Table 2. Continued

Compound Structure Polymers (Drug Load, %) Method Selection Criteria Performance Reference

CAP and PVAP (33%, Ultra-rapid Dissolution, rat PK CAP provided greater degree and 123
50%, 67%) freezing extent of supersaturation in
dissolution. The performance order
on drug load was 33%>50%>67% in
both polymers. Only 33% in CAP
was dosed in rats, which doubled
exposure as compared with
R
Sporanox pellets.
HPMC 2910, Eudragit HME Dissolution, clinical Eudragit E100, mixture of Eudragit 119
E100, mixture of E100-PVPVA64 released drug
Eudragit instantaneously, whereas HPMC
E100-PVPVA64 at released much slower in dissolution.
70/30 (40%) However, HPMC performed the best

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015


in clinical.
HPMC E3, E50, F50, and Solvent shift Precipitation inhibition, HPMCAS family provided greater 29
HPMCAS-L, M, H stabilization of supersaturation,
(10,000x of equilibrium and the stabilization within each
solubility) family was directly related to the
number of hydrophobic functional
groups: HPMCAS H>ML, HPMC
E50>F50E50.
HPMCAS-L, M, H, and KSD Dissolution, rat PK In dissolution: HPMCAS-LM>H. 29
HPMCAS-L, H with C974P reduced AUC for L, but
Carbomer 974P (33%) improved AUC for H. Only 33% in
HPMCAS-L with and without
C974P were dosed in rats, they both
R
performed better than Sporanox
pellets. C974P reduced exposure.
PVP K17, HPMCAS-L SD Molecular mobility HPMCAS-L acted as an 76
(10%) anti-plasticizer of global mobility. It
was substantially more effective
than PVP in inhibiting
crystallization.

Continued

DOI 10.1002/jps.24541
Table 2. Continued

DOI 10.1002/jps.24541
Compound Structure Polymers (Drug Load, %) Method Selection Criteria Performance Reference

LCQ-789, MW 476.9, Tm Eudragit E PO, Eudragit HTP SC, RE Dissolution, rat and dog Eudragit E PO with Cremorphor EL 48
194°C, Tg 105°C, log P L 100, PEG 8000, PK performed better than others in
5.4, pKa 2.5 HPC-L, HPMC, PEG dissolution. It offered exposure
4000, PEG 8000, or similar to lipid-based formulation,
PVP K30 with less variability, and acceptance to
surfactant (10%) toxicology studies.
MK-0364, MW 516.0, Tm HPMCP HP-55, HTP SC, HME, Dissolution, monkey PK PVP/VA 64 with both Tween 80 and 47
104°C, Tg 41°C, log P HPMCAS-L, Eudragit SD Span 80, or VitE TPGS by HME,
6.3, pKa 0.7 L100-55, or PVP/VA 64 HPMC by SD. HMPMCAS-L by SD
with or without offered promising and similar
surfactant (10%) exposure in monkey. HMPMCAS-L
by SD was physically most stable.
Nifedipine, MW 346.3, PVP, HPMC, HPMCAS-M RE Crystallization and phase HPMCAS-M performed the best, 94
Tm 173°C, Tg 45°C, (50%) transition kinetics followed by HPMC, then PVP.
log P 2.3, pKa 3.9
Eudragit L100 and MiCoS XRPD characterization Eudragit L100-55 at 20% drug load, 101
L100-55, HPMCAS-L and HPMCAS-L at 20% and 35%
(20%, 35%, 50%) drug load formed ASDs.
Phenytoin, MW 252.3, Tm PVP K12, K17, VA64, HTP SC, RE Dissolution All three grade of PVP with all three 97
295°C, log P 1.4, pKa PEG6000, Lutrol F68, drug load from HTP performed the
8.3 (acidic) F127, Eugragit E100, best with over 90% phenytoin
Kolliphor HS15, dissoluted within 30 min. The worst
Gekucire 44/14, Brij performance came from the
35, 58, Myrj 49, (10%, combination with Myrj 49. They
20%, 40% in HTP SC) were scaled up by SE and the
(11%, 33% in SE) release profile of laboratory-scale
ASD was similar to that from HTP
films.
Vemurafenib, MW 489.9, HPMCP HP-55, Eudragit MBP Solid stability, All formed ASDs, only Eudragit 124
Tm 272°C, Tg L100-55, HPMCAS-L dissolution, human PK L100-55 and HPMCAS-L ASDs
105°C–107°C, log P (40%) were stable. HPMCAS-L ASD
3.0, neutral provided better dissolution profile
and offered fivefold increased
exposure from crystalline form.

Tg and Tm are from published literature; log P and pKa are from literature or calculated using ACD v12 (Advanced Chemistry Development, Inc.).
PVAP, polyvinyl acetate phthalate; SC, solvent casting; TK, toxicokinetics.
REVIEW

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015


3249
3250 REVIEW

the dose requirement.24,125–127 Given time, the amorphous drug formulations in late-stage development and marketed prod-
in these ASDs will ultimately revert to a crystalline form. Some ucts is growing because of the ASD properties that are well
ASDs, however, have successfully advanced to commercial prod- suited for preparation of conventional oral dosage forms, such
ucts and many more are in the pipeline because not only the as tablets, and capsules.129
bioavailability is optimized, but also the requisite pharmaceu- In one case, a novel ASD technology was employed to rescue
tical stability is achieved when the polymer and manufacturing vemurafenib clinical trials.13 When this new molecular entity
conditions are carefully selected. (NME) was brought to clinical trials, investigators were excited
In an attempt to improve ASD physical stability, increasing by this breakthrough for treatment of late-stage melanoma pa-
the dispersion’s Tg has been the focus of many studies.76 Before tients. At the end of Phase I, the trial had to be halted because
the introduction of more powerful analytical techniques to ana- of low absorption from capsules that were formulated from the
lyze phase separation in ASD, a distinctive single Tg was widely crystalline form of this drug. Patients in the highest dose regi-
accepted as reliable proof of the homogeneity of the amorphous men needed to take 1600 mg vemurafenib, which required in-
mixture at the molecular level.127 In recent years, the interac- gesting 32 capsules.130 The reformulation of vemurafenib suc-
tions between the drug and polymer in the mixture has been ceeded with the development of MBP technology, which led to
much more thoroughly examined and better understood with manufacture this compound as a stable ASD. With more than
the availability of instruments such as ssNMR, DDS, Raman, fivefold increased exposure in the ASD, the dose was reduced
synchrotron radiation, high-speed DSC, and variable tempera- to four tablets twice a day with each tablet containing 240 mg
R
ture XRPD. Besides miscibility between the drug and the poly- vemurafenib (Zelboraf ).124,131 Furthermore, using a less ther-
mer, many other mechanisms have been proposed to explain mally stressed nonsolvent process, KSD technology recently
how the amorphous form of a drug is stabilized by a polymer demonstrated the feasibility to manufacture this drug as an
in ASDs. In some studies, the polymer was found to stabilize ASD with improved efficiency and reduced cost.132 With ASD
the drug in ASD by modifying its "- or $-relaxation (or both technology, the clinical trials were resumed, and the bench to
simultaneously) and consequently delay crystallization onset bedside translation of this breakthrough medicine was realized
time or decrease crystallization rate constant.76 to save the lives of melanoma patients.
In addition to temperature, moisture is another factor that While new therapeutic options can be brought to patients
has profound impact on the stability of ASD. Water is recog- with the application of ASD in NME formulations, the economic
nized as the unavoidable third component in hydrophilic poly- potential of existing drugs can also be improved with applica-
meric dispersion systems at ambient condition.127 With consid- tion of this technology to their reformulation. Besides oral de-
eration of the impact on amorphous drug "- and $-relaxations livery, ASD has also been exploited in injectable formulations.
from both temperature and humidity, the prediction of long- The most frequent applications in injectable formulation are to
term stability (up to 17 months) was achieved and verified by a modify the release profiles of existing drugs. Modified release
statistical method developed using mid-term stability data (up profiles can be realized by forming ASD in microspheres using
to 3 months) from normal and stressed conditions with acceler- SD.133–135 ASD can also be applied in injectable formulations
R
ated temperatures and elevated relative humidities.53 to improve dissolution. Abraxane is an injectable ASD dosage
form that was approved by US FDA and EU in 2005 and 2008,
Generating and Maintaining Supersaturation respectively. This product is supplied as a lyophilized powder
in which amorphous paclitaxel forms a solid dispersion with
Many factors that contribute to the stabilization of the drug in
human albumin.136,137
the solid state of the ASD likely also play a role to maintain
There are increasing numbers of products in pharmaceutical
supersaturation in the solution.
pipelines that are formulated with amorphous solid dispersed
The understanding of polymer interactions with amorphous TM
APIs. A query of the PharmaCircle database138 in March 2015
solids in an aqueous medium to help generate and maintain
revealed 76 products in the pipeline that are formulated as
supersaturated concentration is also improving.93,94,128 Upon
ASDs. These data show that amorphous dispersions are uti-
contact of the ASD with aqueous media, some polymers pro-
lized in NMEs as well as new formulations for lifecycle man-
tect the amorphous solid drug by impeding its crystallization
agement. Of the 27 globally marketed amorphous dispersion
and some polymers dissolve rapidly to increase media viscosity
products, nine were in NME submissions. Some examples of
or interfere the nucleation of the dissolved drug to prevent it
the marketed ASD products used for oral administration are
from precipitating out in the supersaturated solution. In some
listed in Table 3. The FDA and EU approval dates are included
cases, solid polymers swell to facilitate water uptake and wet
when available.
the API. In other cases, polymers dissolve and form micro- or
Even though the confirmed products in research and preclin-
nanoparticles to facilitate fast dissolution of the API. Disso-
ical is only 33 in this search, the actual number of preclinical
lution of a compound from an ASD to generate and maintain
products employing ASDs is likely much higher as the major-
a supersaturated solution is a complicated process. To simplify
ity of studies in pharmaceutical research are not published.
the process of understanding how polymers perform in aqueous
The search also revealed that Bend Research Inc. alone had
media, neat amorphous compounds have been used to evaluate
manufactured over 350 compounds in ASDs for pharmaceuti-
polymer effects by dissolving polymers in the media prior to
cal companies. Among these, over 50 programs have advanced
addition of the amorphous compound.
to clinical trials.
Product and Pipeline
Challenges and Remaining Questions
In addition to the improved dissolution kinetics, flexibility in
dosing, and acceptance in toxicology studies in the discov- As the improved bioavailability using ASD stems from the
ery and early development stages, the number of ASD-based higher energy of the amorphous form, a caveat of this drug

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015 DOI 10.1002/jps.24541


DOI 10.1002/jps.24541
TM
Table 3. Examples of Marketed Amorphous Dispersion Products for Oral Delivery (Source: PharmaCircle )

Product ASD Dosage Therapeutic Formulation


Name Molecule NME/NF WS BCS Polymer Method Form Category FDA EU Patent Company

Advagraf/Astagraf XL Tacrolimus NF ins II HPMC WG Capsule Organ trans. 2013 2007 US6440458 Astellas
Certican/Zortress Everolimus NF vss III HPMC CP Tablet Organ trans. 2010 2003 US6004973 Novartis
Cesamet capsules Nabilone NME vss II PVP SE Capsule Cancer, chemo se 1985 2009 US4087545 Valeant
Fenoglide Fenofibrate NF ins II PEG6000/ CA, SD Tablet Hyperlipidemia 2007 nd US7658944 Santarus
Poloxamer 188 Veloxis
Gris-Peg Griseofulvin NF ins IV PEG 6000 Melt Tablet Infections, fungal 1975 nd nd Pedinol
Incivek Telaprevir NME ins II HPMCAS SD Tablet Infections, hepatitis C 2011 2011 US8431615 Vertex
Intelence Etravirine NME ins IV HPMC HME Tablet Infections, HIV/AIDS 2008 2008 US7887845 Johnson &
Johnson
Isoptin SR Verapamil HCl NF s II HPC/HPMC HME Tablet Hypertension 1997 1986 nd Abbott
Kaletra Ritonavir/ lopinavir NF ins IV PVP HME Tablet Infections, HIV/AIDS 2005 2006 US7364752 AbbVie
ins II
Kalydeco Ivacaftor NME ins II/IV HPMCAS SD Tablet Cystic fibrosis 2012 2012 US8410274 Vertex
Lozanoc Itraconazole NF ins IV HPMCP SD Capsule Infections, fungal nd 2012 US6881745 Mayne
Mesulid fast Nimesulide NF ins II $CD Mech. Tablet Pain nd nd nd Novartis
Norvir Ritonavir NF ins IV PVP HME Tablet Infections, HIV/AIDS 2010 2010 US7364752 AbbVie
Noxafil Posaconazole NF ins II HPMCAS SD, HME Tablet Infections, fungal 2013 2014 US20110123627 Merck
Onmel Itraconazole NF ins IV HPMC HME Tablet Infections, OM 2010 ND US7081255 Merz
Prograf Tacrolimus NME ins II HPMC KDS Capsule Organ trans. 1994 2006 EP0240773 Astellas
Rezulin Troglitazone NME ins II HPMC HME Tablet Diabetes 1997 nd nd Pfizer
Sporanox Itraconazole NF ins IV HPMC SD Capsule Infections, fungal 1992 nd US5633015 Janssen
Thomaflex Meltrex Ibuprofen NF vss II Nd HME Tablet Pain nd 2005 nd Boehringer-
Ingelheim
Zelboraf Vemurafenib NME ins IV HPMCAS MBP Tablet Cancer 2011 2012 nd Roche

CA, controlled agglomeration; CD, cyclodextrin; ins, practically insoluble; KDS, kneading, drying, and sizing; Mech., mechanical; nd, no data; NF, new formulation for existing drug; OM, onychomycosis; s,
soluble; se, side effects; trans., transplantation; vss, very slightly soluble; WG, wet granulation; WS, water solubility.
REVIEW

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015


3251
3252 REVIEW

delivery system is that this higher energy state poses chal- screening of the polymer is better performed using the man-
lenges in manufacture, shipping, storage, and dissolution. Al- ufacturing method that ultimately will be used to produce
though understanding of this drug delivery system has greatly the bulk material for preclinical or clinical development.92,120
improved in recent years, there are many questions that re- In recent years, bench-top spray dryers have often been
main. Understanding the molecular interactions between the used to prepare spray-dried ASD and their appearance in
drug and the polymer carrier in the solid state of the ASD and literature demonstrates their versatility in aiding candi-
in supersaturated solution still need to be further elucidated date selection in early research and downstream formulation
to enhance the predictability of ASD physical and chemical development.27,52,106,112,139
stability and in vivo performance. Further basic research will
certainly boost the applications of ASDs to new levels, and po- Is the Dissolution Method Biorelevant Enough?
tentially enable previously undevelopable drug candidates to
The selection of appropriate dissolution conditions is crucial for
benefit patients.
the successful in vitro evaluation of an ASD formulation. It is
well recognized that the in vitro–in vivo correlation is far from
Can a Polymer be Selected Without Making an ASD? perfect for this type of formulation.114 In the survey performed
by Newman et al.,24 the relationship between in vitro dissolu-
Polymer selection has been thoroughly discussed in both the-
tion and in vivo bioavailability was reported for 78% of the 40
oretical predictions and experimental screening. Although the
studies examined. In 22% of these studies, the in vitro disso-
physical stability of the solid-state ASD can be predicted by
lution failed to offer insight into the in vivo performance. The
theoretical calculations, the performance of ASD still largely
authors systemically analyzed how the dissolution results are
requires experimental evaluation.
influenced by factors including physicochemical properties of
High-throughput solvent casting is the method of choice of
the compound, particle size of the ASD, composition and pH of
some major pharmaceutical companies and it frequently pro-
the dissolution media, selection of dissolution vessel, dissolu-
vides predictable guidance to select an ASD.48,51,98 Instrumen-
tion volume, agitation rate, choice of sink to nonsink conditions,
tation limitations persist as many laboratories are not equipped
and consideration of dose to solubility ratio.24
with automated systems. In addition, when the preparation is
Sample preparation is critical for dissolution studies of this
scaled from HTP screening to laboratory-scale production, the
type of formulation. They should be designed based on the in
properties of the product sometimes cannot be reproduced be-
vivo dosing protocol. If the particle size of the dispersion is to
cause of the change in thermal history. In scale-up, the process
be controlled for in vivo dosing, for example, sieved to a more
can be altered from film casting to SD or melt extrusion or other
defined particle size range, it should be controlled in the disso-
techniques. The mechanism varies from solvent-based to melt-
lution too. Although solid dosage formulations such as capsules
based, or to mechanical-based or vice versa.30,114 When an ASD
are preferred in preclinical studies to facilitate animal dos-
of felodipine was prepared using PVP K30 or HPMCAS-L as
ing, ASDs are frequently dosed as suspensions in preclinical
the carrier, the drug was released faster from the spray-dried
studies, especially when the carrier is an enteric polymer. If a
product than from hot-melt extruded product.120 As the crys-
suspension of the dispersion is planned to be dosed sometime
tallization of felodipine happens rapidly in the solid once the
after preparation, for example, 1 day or 1 week after prepara-
ASD contacts the aqueous medium, the thermal history has
tion, the prepared sample should be kept for the same length
important implication on how the polymer interacts with the
of time prior to evaluation via dissolution testing. The role of
compound.128
the vehicle in this type of suspensions is to maintain the solid
Although the stability of the ASD is important, improved
in amorphous form. The physical form of the ASD must also
bioavailability is the key criterion of an ASD. The in vivo per-
be evaluated to ensure there is no change after the sample is
formance is determined by many factors, including crystalliza-
prepared.
tion tendency of the compound in solid form upon contacting
Because of the metastable nature of this type of dispersion,
the GI fluid, dissolution of the amorphous drug, dissolution of
it usually does not afford the luxury to prepare a single sample
the polymer, interaction of the drug–polymer in both solid and
for a 2-week toxicology study when the formulation is a suspen-
solution, precipitation of the supersaturated drug in its crys-
sion. The need for extemporaneous preparation for each dose is
talline form, and so on. When the performance is dominated by
common. Stability of at least 2 h is usually preferred, although
the polymer’s ability to generate and maintain supersaturation,
some institutes accept half-hour stability.48 If the suspension
such as in the case of BMS-A, the solvent shift method can give
stability is unknown, ASD material can be dispersed into the
a reliable prediction.112 When the ASD performance is mainly
dosing vehicle immediately prior to dosing.122 The dose to media
contributed by the interaction between drug and polymer to
volume ratio should also be carefully selected, as precipitation
generate and hold the supersaturation, the ASD may have to
from supersaturation is a concentration-driven phenomenon.
be prepared for screening. When the kinetics during ASD man-
ufacture do not significantly contribute to the properties of the
Other Considerations in Dissolution
ASD, the polymer can be screened without manufacturing the
ASD or the ASD produced by any manufacturing process may The vehicle composition of formulation is important. Tween 80
be used.51,121 A study of griseofulvin on HPMCAS-M matrix at a concentration of 0.2%–0.5% is routinely used as a wet-
using small-scale bench processes, namely, fast evaporation, ting agent in suspensions of crystalline material. However, it
lyophilization, and SD, demonstrated that each of these tech- reduced the drug release and led to precipitation in a short pe-
niques is reliable, comparable, and suitable to generate ASD riod of time in the case of an LCQ789 ASD suspension,48 which
material without large investment in time and API quantity.121 is consistent with the authors’ observations with other research
However, when the thermal history of the manufacturing compounds (unpublished data). In another study, Tween 80
process significantly contributes to the ASD properties, the at a concentration of 10% was added in an ASD suspension

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015 DOI 10.1002/jps.24541


REVIEW 3253

formulation because it provided significant solubility improve- Other animal species, such as rats, also were reported to
ment for the neat crystalline compound. It was expected that have pH variability in the duodenum and jejunum and this
Tween 80 would act like parachute to delay precipitation from can be further impacted by food intake.29 Although the degree
the supersaturated solution of ASD. Surprisingly, the solid dis- of pH variation is less as compared with that of monkeys, high
persion crystallized after 3 h in the media with Tween 80, variability of exposure was still observed when a pH-dependent
whereas it was stable for more than 6 h when Tween 80 was soluble polymer was used in the ASD.29,122,145 The reproducibil-
not present.5 ity was excellent, when fed dogs were dosed with powder in hard
When an ASD is dosed at low concentrations, suspending gelatin capsules (with 2% disintegrant added) using different
agents, such as methylcellulose, may need to be added.5 How- batches of ASD from MBP.13
ever, when ASD is dosed at higher concentrations, water or Regardless of the potential exposure variation, pH-
buffer without suspending agent may be used as the amount dependent polymers are frequently selected as the carrier be-
of polymer released from the ASD can serve as a suspension cause they provide the exposure improvement in much higher
agent. Water was selected to suspend the LCQ789 ASD, which magnitudes and offer the benefit of targeted delivery or delayed
facilitated formulations of 10, 50, and 120 mg/mL of ASD.48 Tmax . The exposure variability and higher probability of food ef-
When the compound is pH sensitive or the polymer solubil- fect need to be included in the considerations of the dissolution
ity is pH dependent, the pH of the suspension vehicle and the method for preclinical and clinical study designs to fully under-
buffer composition should be carefully selected. The prepara- stand how they can affect the study outcomes and how they can
tion should avoid energetic mixing as it can accelerate reversion be maximally mitigated and controlled.
to a crystalline form. The powder can be effectively wetted first One of the major benefits of using ASD for preclinical studies
by adding a minimal amount of suspension vehicle and stirring is that it can provide much higher doses in toxicology dose esca-
gently with a spatula. lation studies because of the higher achievable drug exposure,
The dissolution temperature of an ASD formulation is im- inert nature of the polymer and the dosing flexibility of sus-
portant too. It should be set close to physiological temperature. pension formulation. ASDs can be dosed as simple suspensions
Although equilibrium solubility increases with elevated tem- in preclinical studies and early clinical trials, can be formu-
perature and the solubility difference between 25°C and 37°C is lated and encapsulated as powder in capsule or compressed as
usually minimal,140,141 the solubility of a neat amorphous com- tablets in formulation development, and can provide a clear
pound or ASD may decrease with increased medium temper- development pathway to a commercial product.
ature because crystallization onset may be earlier, crystalliza- The optimal drug loading in an ASD is usually in the range
tion rate may be faster, amorphous material agglomerates may of 10%–30% as further increasing drug loading can jeopardize
be more extensive, and crystalline precipitation from supersat- manufacturing stability and shelf life, and also can negatively
uration may also occur sooner.128 Therefore, dissolution at a impact its dissolution performance.93,121,123 When a high dose is
biologically relevant temperature is necessary for ASD-based projected for clinical usage, pill burden can impact clinical ac-
formulations, whereas dissolution at room temperature can be ceptability. The maximum daily doses of acceptable excipients
acceptable for dosage forms containing crystalline phases. should also be considered. In a study of berberine, the selected
When food effect is evaluated, the consideration should be ASD contained a 1:1:6 ratio of berberine–sodium caprate–PEG
not only the bile salts composition of the media, but also how 6000.95 On the basis of bioavailability and antidiabetic efficacy
the food affects the pH, fluid volume, and the residence time in in a type 2 diabetic rat model, it was proposed that this ASD
each segment of the GI tract. could decrease the berberine clinical dose to one quarter of 0.9–
1.5 g per day, which would require 1.8–3.0 g ASD per day. This
amount of ASD would contain 0.2–0.4 g sodium caprate per day.
It is important to know whether the total amount of inactive
Can Preclinical Results Translate to Clinical?
ingredient dosed is toxicologically and clinically acceptable.
During preclinical studies, the selection of animal species is
clearly critical as it determines if the results can be trans-
ASD Selection Road Map
lated to the clinical studies. Although disease relevance is of-
ten the focus of the animal species selection, the PK profile The physicochemical properties of the drug substance deter-
of a compound is greatly influenced by the physiology and mine whether ASD can be a choice to improve its bioavailabil-
anatomy of the animal. As summarized in the survey by New- ity. A compound with low Tm and high Tg would have fewer
man et al.,24 dog and rat are the most favorable choice when obstacles to convert to an amorphous phase and less thermo-
ASDs are dosed. Nonhuman primates have been used in stud- dynamic driving force for the amorphous form to revert back to
ies of ASDs and they are regarded as useful models in drug its crystalline form. A good glass former with low crystallizing
delivery with regard to gastric pH and emptying time, GI ag- tendency is more suitable for forming an ASD.
itation intensity, and intestinal transit time.27,142,143 However, The selection of the polymer is largely governed by the solu-
large exposure variability could limit their use when the com- bility of the drug in the polymer. Many efforts have been made
pound is ionizable or when a pH-dependent polymer is used as for a theoretical prediction. Limited by the inadequate under-
the carrier. The cause of the variability is often the consump- standing of the mechanisms involved with stabilization, disso-
tion of a meal that has strong impact on their GI pH profiles.144 lution, and crystallization in supersaturated solutions of ASD,
In addition, primates are not good models for fed-state studies the selection of the polymer and the optimization of the drug
because they eat when they choose and often do not eat at the load in the ASD remains largely empirical and relies mainly on
moment food is provided. The duration of gastric pH elevation experimental screening.2,76
after ingestion of a meal varies considerably in primates, and When the API quantity is limited, the selection of polymer is
intestinal pH is more basic in primates than in humans.27 traditionally screened using solvent quench or SE methods.89,92

DOI 10.1002/jps.24541 He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015


3254 REVIEW

The evolution of this field has seen the recent introduction of other considerations should include the safety of the polymer
new methods such as single droplet drying and miniaturized in animal and humans, suitable properties for the selected
coprecipitation screening (MiCoS). With the availability of au- manufacturing processing method, the release profile in disso-
tomated systems, some of these screening techniques can be lution, and stability in both solid and supersaturated solutions.
adapted to a HTP processes as depicted in Figure 2. These In current pharmaceutical industry practice, ASDs are most
methods can be used, variously, to screen for the feasibility of commonly prepared from SD in research and HME is more
forming an ASD, the stability of the ASD, and the optimum preferable in development because of the factors discussed in
ASD drug loading as evaluated by stability and dissolution. the introduction section. An ASD formulation approach would
Miniaturized spray dryers also have enabled screening with likely not be appropriate when developing a conventional for-
milligram quantities of API. Many studies have demonstrated mulation with acceptable performance is possible. In instances
that ASD generated by miniaturized spray driers can be repro- when an ASD is selected for discovery stage research and a
duced at larger development scale. At present, the application conventional formulation becomes feasible at the clinical stage,
of nonsolvent process ASD in research and early development the transition should be considered as early as possible to avoid
remains challenging because of the constraints of the amount expensive bridging studies in later-stage clinical trials.
of drug required for the evaluation. The currently available Drug loading is often optimized after the ASD is selected as
batch mode bench top HME instrumentation requires a mini- the delivery technology. The optimization is a balanced consid-
mum of 5 g of drug–polymer mixture, whereas more material is eration of the solid stability, dissolution performance, in vivo
required in the continuous process model. When a technology exposure, and pill burden in the final dosage form.
breakthrough introduces a scalable miniaturized solvent-free As evident from the literature cited throughout this review
instrument, such as HME or KSD, to enable the screening of and the data presented in the product and pipeline sections,
solvent free ASDs using milligrams of API, the application of the technologies available for screening ASDs have matured
pharmaceutical ASDs will be further accelerated. tremendously at this time. Importantly, the processes for large-
Once the compound is identified as an ASD candidate, en- scale manufacturing of ASD are more established. FDA ap-
abling formulations by other approaches cannot be formulated proval of these novel drug products also seems simpler because
or the benefit of developing ASD formulation is viewed as the regulators are more comfortable with such formulations.
overweighing the benefits of other formulations, the method Companies are seizing the advantages of the unique intellec-
of screening might be governed by the practical factors, such tual property positions accessible through ASDs. The pharma-
as the amount of API available for feasibility testing, in-house ceutical industry has embraced the idea that the applications of
expertise, and access of instruments. Being a good ASD candi- ASDs are not limited to NMEs, but also been to lifecycle man-
date does not warrant the compound to stand all ASD process- agement of existing medications.17 It is now clear that ASD
ing options. Compounds, such as vemurafenib, cannot stand technology has become an important drug delivery method for
HME because of its high Tm and thermal instability and can- pharmaceutical scientists.
not be manufactured by SD because its low solubility in volatile
solvents and water. Some research compounds can be manu-
factured using SD with relative low solubility in volatile sol-
CONCLUSIONS
vents because the amount of ASD needed at this stage is
small. However, after the ASD is proved to be the right con- The successful formulation of an ASD is dependent on its sta-
cept for the compound and a larger amount of ASD is needed in bility in the solid state and its performance after dosing. The
the later stage of the pipeline, the amount of solvent needed manufacturing challenges and the phase stability upon storage
in SD can be a restraint and another process that has ac- are hurdles to the acceptance and wider application of ASD. In
ceptable processability at scaled-up setting may have to be the last two decades, increased efforts from academic and in-
explored. Shelf life in pharmaceutics is a relevant term. Al- dustrial researchers have pushed the understanding to a new
though a few months stability is usually sufficient for research- level regarding the fundamental driving forces of (1) how ASD
stage compounds up to GLP toxicology studies, a longer time works to improve exposure, (2) what are the thermodynamic
frame shelf life will be required for clinical studies that can and kinetic factors as well as molecular interaction between
add another factor for processing selection. The reliable pre- drug and polymer for its phase stability in solid state, and (3)
diction of long-term stability from short-term data can help how the supersaturated solution is generated and maintained
ensure confidence in the compound transition from preclini- in the GI tract.
cal to clinical. Robust batch to batch physical quality repro- Manufacturing challenges are also better controlled. With
ducibility of ASD is also important in the application of this currently available instruments, in particular bench-top spray
technology. dryers, it is possible to prepare ASD with minimal amount of
The emergence of new types of modified polymers, such as material to perform a quick bioavailability study, when other
HPMCAS and Soluplus, offer better stability and dissolution approaches are exhausted for a poorly water-soluble compound
profiles for some drugs. The decision to employ a new unap- in the research or early development stages. Although the
proved excipient is not only a pharmacologic safety risk, but choice of technique in marketed product is dominated by HME,
also a regulatory and business decision. The pioneers using other techniques such as SD, CP, MBP, SE, mechanical milling,
new polymers for ASDs have a strong advantage in terms of and kneading are also being used.
patent protection. However, in order to gain regulatory ap- Despite the fact that ASD is usually the last choice of phar-
proval, there is a daunting amount of work and expense to maceutical scientists because of the challenges in manufactur-
introduce new excipients. Polymer carrier selection criteria ing and physical stability, it has been demonstrated in numer-
were well discussed in the review article by Janssens and Van ous cases that it can be a very effective and powerful approach
den Mooter.8 Although the stability of the ASD is important, to improve exposure and facilitate project progression from lead

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015 DOI 10.1002/jps.24541


REVIEW 3255

optimization, to candidate selection, to full development and throughput optimization in support of drug discovery. Pharm Dev Tech-
into a commercial product. Improvement in the understanding nol 18(1):121–129.
of both the solid-state stability of ASDs and the mechanism 16. Gaspar F. 2014. Spray drying in the pharmaceutical industry. Eur
of generating and maintaining supersaturation have led the Pharm Rev 19(5):45–48.
17. Siew A. 2014. Solving poor solubility with amorphous solid disper-
paradigm shift from avoiding amorphous forms in drug prod-
sions. Pharm Tech 38(10).
ucts because of physical and chemical instability to actively
18. Sekiguchi K, Obi N. 1961. Studies on absorption of eutectic mixture.
utilizing ASDs to enhance drug absorption by taking advan- I. A comparison of the behavior of eutectic mixture of sulfathiazole and
tages of the powerful solubility enhancement that is intrinsic that of ordinary sulfathiazole in man. Chem Pharm Bull 9(11):866–
to the technology. 872.
19. Chiou WL, Riegelman S. 1971. Pharmaceutical applications of solid
dispersion systems. J Pharm Sci 60(9):1281–1302.
ACKNOWLEDGMENT 20. Goldberg AH, Gibaldi M, Kanig JL. 1966. Increasing dissolution
rates and gastrointestinal absorption of drugs via solid solutions and
The authors gratefully appreciate Dr. Harvey Lieberman and eutectic mixtures II. Experimental evaluation of eutectic mixture:
Dr. Donglai Yang for their discussions and prereview, and sin- Urea-acetaminophen system. J Pharm Sci 55(5):482–487.
cerely thank Dr. Ed Orton for help in editing this manuscript. 21. Goldberg AH, Gibaldi M, Kanig JL. 1966. Increasing dissolution
rates and gastrointestinal absorption of drugs via solid solutions and
eutectic mixtures III. Experimental evaluation of griseofulvin-succinic
acid solution. J Pharm Sci 55(5):487–492.
REFERENCES
22. Serajuddin ATM. 1999. Solid dispersion of poorly water-soluble
1. Di L, Fish PV, Mano T. 2012. Bridging solubility between drug dis- drugs: Early promises, subsequent problems, and recent break-
covery and development. Drug Discov Today 17:486–495. throughs. J Pharm Sci 88(10):1058–1066.
2. Williams HD, Trevaskis NL, Charman SA, Shanker RM, Charman 23. Leuner C, Dressman J. 2000. Improving drug solubility for oral
WN, Pouton CW, Porter CJ. 2013. Strategies to address low drug solu- delivery using solid dispersion. Eur J Pharm Biopharm Sci 50(1):47–
bility in discovery and development. Pharmacol Rev 65(1):315–499. 60.
3. Neervannan S. 2006. Preclinical formulations for discovery and tox- 24. Newman A, Knipp G, Zografi G. 2012. Assessing the performance
icology: Physicochemical challenges. Expert Opin Drug Metab Toxicol of amorphous solid dispersions. J Pharm Sci 101(4):1355–1377.
2(5):715–731. 25. Taylor L, Hancock B. 2014. George Zografi and the science of solids
4. Maas J, Kamm W, Hauck G. 2007. An integrated early formulation and surfaces. J Pharm Sci 103(9):2592–2594.
strategy—From hit evaluation to preclinical candidate profiling. Eur J 26. Palucki M, Higgins J, Kwong E, Templeton A. 2010. Strategies at
Pharm Biopharm 66(1):1–10. the interface of drug discovery and development: Early optimization of
5. Lohani S, Cooper H, Jin X, Nissley BP, Manser K, Rakes LH, the solid state phase and preclinical toxicology formulation for potential
Cummings JJ, Fauty SE, Bak A. 2014. Physicochemical properties, drug candidates. J Med Chem 53:5897–5905.
form, and formulation selection strategy for a biopharmaceutical clas- 27. Chen XQ, Stefanski K, Shen H, Huang C, Caporuscio C, Yang W,
sification system class II preclinical drug candidate. J Pharm Sci Lam P, Su C, Gudmundsson O, Hageman M. 2014. Oral delivery of
103(10):3007–3021. highly lipophilic poorly water-soluble drugs: Spray-dried dispersions to
6. Stegemann S, Leveiller F, Franchi D, de Jong H, Lindén H. 2007. improve oral absorption and enable high-dose toxicology studies of a
When poor solubility becomes an issue: From early stage to proof of P2Y1 antagonist. J Pharm Sci 103(12):3924–3931.
concept. Eur J Pharm Sci 31(5):249–261. 28. Konno K, Taylor LS. 2006. Influence of different polymers on the
7. Yalkowsky SH. 2012. Perspective on improving passive human in- crystallization tendency of molecularly dispersed amorphous felodip-
testinal absorption. J Pharm Sci 101(9):3047–3050. ine. J Pharm Sci 95(12):2692–2705.
8. Janssens S, Van den Mooter G. 2009. Physical chemistry of solid 29. DiNunzio JC, Hughey JR, Brough C, Miller DA, Williams RO 3rd,
dispersions. J Pharm Pharmacol 61(12):1571–1586. McGinity JW. 2010. Production of advanced solid dispersions for en-
9. Padden, BE, Miller, JM, Robbins T, Zocharski PD, Prasad L, Spence hanced bioavailability of itraconazole using KinetiSol dispersing. Drug
JK, LaFountaine J. 2011. Amorphous solid dispersions as enabling Dev Ind Pharm 36(9):1064–1078.
formulations for discovery and early development. Am Pharm Rev 30. Janssens S, De Zeure A, Paudel A, Van Humbeeck J, Rom-
14(1):66, 68–70, 72–73. baut P, Van den Mooter G. 2010. Influence of preparation meth-
10. Das SK, Roy S, Kalimuthu Y, Khanam J, Nanda A. 2011. Solid ods on solid state supersaturation of amorphous solid dispersions: A
dispersions: An approach to enhance the bioavailability of poorly water- case study with itraconazole and Eudragit E100. Pharm Res 27(5):
soluble drugs. Int J Pharmacol Pharma Technol 1(1):37–46. 775–785.
11. Sareen S, Mathew G, Joseph L. 2012. Improvement in solubility 31. Frank KJ, Westedt U, Rosenblatt KM, Hoelig P, Rosenberg J,
of poor water-soluble drugs by solid dispersion. Int J Pharm Investig Maegerlein M, Fricker G, Brandl M. 2014. What is the mecha-
2(1):12–17. nism behind increased permeation rate of a poorly soluble drug from
12. Sharma M, Garg R, Gupta GD. 2013. Formulation and evaluation aqueous dispersions of an amorphous solid dispersion? J Pharm Sci
of solid dispersion of atorvastatin calcium. J Pharm Sci Innovation 103(6):1779–1786.
2(4):73–81. 32. Lian X, Dong J, Zhang J, Teng Y, Lin Q, Fu Y, Gong T. 2014. Solu-
13. Shah N, Sandhu H, Phuapradit W, Pinal R, Iyer R, Albano A, plus based 9-nitrocamptothecin solid dispersion for peroral adminis-
Chatterji A, Anand S, Choi DS, Tang K, Tian H, Chokshi H, Singhal D, tration: Preparation, characterization, in vitro and in vivo evaluation.
Malick W. 2012. Development of novel microprecipitated bulk powder Int J Pharm 477(1–2):399–407.
(MBP) technology for manufacturing stable amorphous formulations of 33. Guzmán H, Tawa M, Zhang Z, Ratanabanangkoon P, Shaw P,
poorly soluble drugs. Int J Pharm 438(1–2):53–60. Mustonen P, Gardner C, Chen H, Moreau J, Almarsson O, Remenar
14. Patel RC, Masnoon S, Patel MM, Patel NM. 2009. Formulation J. 2004. A “spring and parachute” approach to designing solid cele-
strategies for improving drug solubility using solid dispersions. Pharm coxib formulations having enhanced oral absorption. AAPS J 6:Abstract
Rev 7(6):1918–1922. T2189.
15. Ormes JD, Zhang D, Chen AM, Hou S, Krueger D, Nelson T, 34. Guzmán HR, Tawa M, Zhang Z, Ratanabanang-koon P, Shaw P,
Templeton A. 2013. Design of experiments utilization to map the Gardner CR, Chen H, Moreau J, Almarsson O, Remenar JF. 2007.
processing capabilities of a micro-spray dryer: Particle design and Combined use of crystalline salt forms and precipitation inhibitors

DOI 10.1002/jps.24541 He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015


3256 REVIEW

to improve oral absorption of celecoxib from solid oral formulations. 54. Jacobs IC, Higgins JD, Guillot M, Franson NM, Rocco WL,
J Pharm Sci 96(10):2686–2702. Abu-Izza, KA. 2007. Preparation of amorphous solid dispersions of 7-
35. Hancock BC, Parks M. 2000. What is the true solubility advantage chloro-N,N,5-trimethyl-4-oxo-3-phenyl-3,5-dihydro-4H-pyridazino[4,5-
for amorphous pharmaceuticals? Pharm Res 17(4):397–404. b]indole-1-acetamide. Patent WO 2007027494 A2.
36. Percy SR. 1872. Improvement in drying and concentrating liquid 55. Kwong AD, Kauffman RS, Hurter P, Mueller P. 2011. Discovery and
substances by atomizing. Patent US 125406 A development of telaprevir: An NS3–4A protease inhibitor for treating
37. Masters K. 2002. Spray drying in practice, SprayDryConsult Inter- genotype 1 chronic hepatitis C virus. Nat Biotechnol 29(11):993–1003.
national, Denmark. 56. Friesen DT, Shanker R, Crew M, Smithey DT, Curatolo
38. Kornblum SS, Hirschorn JO. 1970. Dissolution of poorly water- WJ, Nightingale JA. 2008. Hydroxypropyl methylcellulose acetate
soluble drugs I: Some physical parameters related to method of mi- succinate-based spray-dried dispersions: An overview. Mol Pharm
cronization and tablet manufacture of a quinazolinone compound. J 5(6):1003–1019.
Pharm Sci 59(5):606–609. 57. Kawakami K, Usui T, Hattori M. 2012. Understanding the glass-
39. Chan HK, Kwok PCL. 2011. Production methods for nanodrug forming ability of active pharmaceutical ingredients for designing su-
particles using the bottom-up approach. Adv Drug Deliv Rev 63: persaturating dosage forms. J Pharm Sci 101(9):3239–3248.
406–416. 58. Zhou D, Zhang GG, Law D, Grant DJ, Schmitt EA. 2002. Physi-
40. Kolašinac N, Kachrimanis K, Djuriš J, Homšek I, Grujić B, Ibrić cal stability of amorphous pharmaceuticals: Importance of configura-
S. 2013. Spray coating as a powerful technique in preparation of solid tional thermodynamic quantities and molecular mobility. J Pharm Sci
dispersions with enhanced desloratadine dissolution rate. Drug Dev 91(8):1863–1872.
Ind Pharm 39(7):1020–1027. 59. Mahlin D, Ponnambalam S, Höckerfelt MH, Bergström CA. 2011.
41. Knopp MM, Olesen NE, Holm P, LÖBmann K, Holm R, Langguth P, Toward in silico prediction of glass-forming ability from molecular
Rades T. 2015. Evaluation of drug–polymer solubility curves through structure alone: A screening tool in early drug development. Mol Pharm
formal statistical analysis: Comparison of preparation techniques. J 8(2):498–506.
Pharm Sci 104(1):44–51. 60. Mahlin D, Bergstrom CA. 2013. Early drug development predictions
42. Ayad MH, Bonnet B, Quinton J, Leigh M, Poli SM. 2013. Amorphous of glass-forming ability and physical stability of drugs. Eur J Pharm
solid dispersion successfully improved oral exposure of ADX71943 Sci 49(2):323–332.
in support of toxicology studies. Drug Dev Ind Pharm 39(9):1300– 61. Amjad A, Alzghoul A, Kaialy W, Mahlin D, Bergstroem CA. 2014.
1305. Computational predictions of glass-forming ability and crystallization
43. Law D, Schmitt EA, Marsh KC, Everitt EA, Wang W, Fort JJ, Krill tendency of drug molecules. Mol Pharm 11(9):3123–3132.
SL, Qiu Y. 2004. Ritonavir-PEG 8000 amorphous solid dispersions: In 62. Trasi NS, Baird JA, Kestur US, Taylor LS. 2014. Factors influenc-
vitro and in vivo evaluations. J Pharm Sci 93(3):563–570. ing crystal growth rates from undercooled liquids of pharmaceutical
44. Liepold B, Jung T, Holig P, Schroeder R, Sever NE, Lafountaine J, compounds. J Phys Chem B 118(33):9974–9982.
Sinclair BD, Gao Y, Wu J, Erickson BK, Kullmann S, Wstedt U, Pauli 63. Hancock BC, Zografi G. 1997. Characteristics and significance of
M, Meitermann T, Koenig R, Thiei M. 2011. Solid compositions of a the amorphous state in pharmaceutical systems. J Pharm Sci 86(1):1–
hepatitis C virus inhibitor. Patent WO 2011156578 A1. 12.
45. Fakes MG, Vakkalagadda BJ, Qian F, Desikan S, Gandhi RB, Lai 64. Baird JA, Van Eerdenbrugh B, Taylor LS. 2010. A classification
C, Hsieh A, Franchini MK, Toale H, Brown J. 2009. Enhancement of system to assess the crystallization tendency of organic molecules from
oral bioavailability of an HIV-attachment inhibitor by nanosizing and undercooled melts. J Pharm Sci 99(9):3787–3806.
amorphous formulation approaches. Int J Pharm 370(1–2):167–174. 65. Kawakami K, Harada T, Miura K, Yoshihashi Y, Yonemochi E,
46. Vickery RD, Stefanski KJ, Su CC, Hageman MJ, Vig BS, Betigeri S. Terada K, Moriyama H. 2014. Relationship between crystallization ten-
2013. Bioavailable compositions of an amorphous solid dispersions of a dencies during cooling from melt and is Periodicalmal storage: Toward a
piperidinyl compound for capsules and tablets. Patent WO 2013013114 general understanding of physical stability of pharmaceutical glasses.
A1. Mol Pharm 11(6):1835–1843.
47. Sotthivirat S, McKelvey C, Moser J, Rege B, Xu W, Zhang D. 2013. 66. Kauzmann W. 1948. The nature of the glassy state and the behavior
Development of amorphous solid dispersion formulations of a poorly of liquids at low temperatures. Chem Rev 115(2):219–256.
water-soluble drug, MK-0364. Int J Pharm 452(1–2):73–81. 67. Beaman RG. 1952. Relation between (apparent) second-order tran-
48. Zheng W, Jain A, Papoutsakis D, Dannenfelser RM, Panicucci R, sition temperature and melting point. J Polym Sci 9(5):470–472.
Garad S. 2012. Selection of oral bioavailability enhancing formulations 68. Boyer RF. 1963. The relation of transition temperatures to chemical
during drug discovery. Drug Dev Ind Pharm 38(2):235–247. structure in high polymers. Rubber Chem Technol 36(5):1303–1421.
49. Ruggeri RB, Magnus-Aryitey G, Shanker RM, Lorenz DA, 69. Akihisa I, Tao Z, Tsuyoshi M. 1989. Aluminum–lanthanum–nickel
Garr CD. 2006. Preparation of substituted benzylamino-1,2,3,4- amorphous alloys with a wide supercooled liquid region. Mater Trans
tetrahydroquinoline derivatives for use as cholesterol lowering agents. 30(12):965–972.
Patent US 20060063803 A1. 70. Wei BC, Wang WH, Xia L, Zhang A, Zhao DQ, Pan MX. 2002.
50. Curatolo WJ, Friesen DT, Sutton SC. 2005. Controlled release Glass transition and thermal stability of hard magnetic bulk NdAlFeCo
dosage forms containing cholesteryl ester transfer protein inhibitors metallic glass. Mater Sci Eng A 334(1–2):307–311.
and immediate release of HMG-CoA reductase inhibitors. Patent WO 71. Zallen R. 1983. The formation of amorphous solids. In The physics
2005011634 A1. of amorphous solids. New York: Wiley, pp 1–22.
51. Chiang PC, Ran Y, Chou KJ, Cui Y, Sambrone A, Chan C, Hart 72. Ping W, Paraska D, Baker R, Harrowell PC, Angell A. 2011.
R. 2012. Evaluation of drug load and polymer by using a 96-well plate Molecular engineering of the glass transition: Glass-forming ability
vacuum dry system for amorphous solid dispersion drug delivery. AAPS across a homologous series of cyclic stilbenes. Mol Pharm 115(16):
Pharm Sci Tech 13(2):713–722. 4696–4702.
52. Cui Y, Chiang PC, Choo EF, Boggs J, Rudolph J, Grina J, 73. Marsac PJ, Li T, Taylor LS. 2009. Estimation of drug–polymer mis-
Wenglowsky S, Ran Y. 2013. Systemic in vitro and in vivo evaluation cibility and solubility in amorphous solid dispersions using experimen-
for determining the feasibility of making an amorphous solid disper- tally determined interaction parameters. Pharm Res 26(1):139–151.
sion of a B-Raf (rapidly accelerated fibrosarcoma) inhibitor. Int J Pharm 74. Guo Y, Shalaev E, Smith S. 2013. Solid-state analysis and amor-
454(1):241–248. phous dispersions in assessing the physical stability of pharmaceutical
53. Greco S, Authelin JR, Leveder C, Segalini A. 2012. A practical formulations. TrAC Trend Anal Chem 49:137–144.
method to predict physical stability of amorphous solid dispersions. 75. Paudel A, Geppi M, Van Den Mooter G. 2014. Structural and dy-
Pharm Res 29(10):2792–2805. namic properties of amorphous solid dispersions: The role of solid-state

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015 DOI 10.1002/jps.24541


REVIEW 3257

nuclear magnetic resonance spectroscopy and relaxometry. J Pharm demonstrates a remarkable hypoglycemic effect via improving its
Sci 103(9):2635–2662. bioavailability. Int J Pharm 467(1–2):50–59.
76. Bhardwaj SP, Arora KK, Kwong E, Templeton A, Clas SD, 96. Lee T, Lee J. 2003. Drug-carrier screening on a chip. Pharm Technol
Suryanarayanan R. 2014. Mechanism of amorphous itraconazole 1:40–48.
stabilization in polymer solid dispersions: Role of molecular mobility. 97. Barillaro V, PaPescarmona PP, Van Speybroeck M, Do Thi T, Van
Mol Pharm 11(11):4228–4237. Humbeeck J, Vermant J, Augustijns P, Martens JA, Van Den Mooter
77. Worku ZA, Aarts J, Singh A, Van den Mooter G. 2014. Drug-polymer G. 2008. High-throughput study of phenytoin solid dispersions: Formu-
miscibility across a spray dryer: A case study of naproxen and micona- lation using an automated solvent casting method, dissolution testing,
zole solid dispersions. Mol Pharm 11(4):1094–1101. and scaling-up. J Comb Chem 10:637–643.
78. Qian F, Huang J, Hussain MA. 2010. Drug-polymer solubility and 98. Mansky P, Dai W, Li S, Pollock-Dove C, Daehne K, Dong L,
miscibility: Stability consideration and practical challenges in amor- Eichenbaum G. 2007. Screening method to identify preclinical liquid
phous solid dispersion development. J Pharm Sci 99(7):2941–2947. and semi-solid formulations for low solubility compounds: Miniaturiza-
79. Flory RJ. 1953. Principles of polymer chemistry. Ithaca, New York: tion and automation of solvent casting and dissolution testing. J Pharm
Cornell University Press. Sci 96(6):1548–1563.
80. Marsac P, Shamblin S, Taylor L. 2006. Theoretical and practical 99. Shanbhag A, Rabel S, Nauka E, Casadevall G, Shivanand P,
approaches for prediction of drug-polymer miscibility and solubility. Eichenbaum G, Mansky P. 2008. Method for screening of solid disper-
Pharm Res 23(10):2417–2426. sion formulations of low-solubility compounds—Miniaturization and
81. Zhao Y, Inbar P, Chokshi HP, Malick AW, Choi DS. 2011. Prediction automation of solvent casting and dissolution testing. Int J Pharm
of the thermal phase diagram of amorphous solid dispersions by Flory- 351(1–2):209–218.
Huggins theory. J Pharm Sci 100(8):3196–3207. 100. Wenglowsky S, Ren L, Ahrendt KA, Laird ER, Aliagas I, Alicke B,
82. Thakral S, Thakral NK. 2013. Prediction of drug-polymer misci- Buckmelter AJ, Choo EF, Dinkel V, Feng B, Gloor SL, Gould SE, Gross
bility through the use of solubility parameter based Flory-Huggins in- S, Gunzner-Toste J, Hansen JD, Lord-Ondash HA, Malesky K, Mathieu
teraction parameter and the experimental validation: PEG as model S, Newhouse B, Raddatz NJ, Ran Y, Rana S, Randolph N, Risom T,
polymer. J Pharm Sci 102(7):2254–2263. Rudolph J, Savage S, Selby LT, Shrag M, Voegtli WC, Wen Z, Willis
83. Huang Y, Dai W. 2014. Fundamental aspects of solid dispersion BS, Woessner RD, Wu WI, Young WB, Grina J. 2011. A selective orally
technology for poorly soluble drugs. Acta Pharm Sin B 4(1):18–25. bioavailability and efficacious pyrazolopyridine inhibitor of V600EB-
84. Donnelly C, Tian Y, Potter C, Jones DS, Andrews GP. 2014. Probing Raf. ACS Med Chem Lett 2(5):342–347.
the effects of experimental conditions on the character of drug-polymer 101. Hu Q, Choi DS, Chokshi H, Shah N, Sandhu H. 2013. Highly
phase diagrams constructed using Flory-Huggins theory. Pharm Res efficient miniaturized coprecipitation screening (MiCoS) for amor-
32(1):167–179. phous solid dispersion formulation development. Int J Pharm 450(1–2):
85. Hildebrand JH, Scott RL. 1950. The solubility of non-electrolytes. 53–62.
3rd ed. New York: Reinhold. 102. Adhikari B, Howes T, Bhandari BR, Truong V. 2000. Experimental
86. Lindvig T, Michelsen ML, Kontogeorgis GM. 2002. A Flory– studies and kinetics of single drop drying and their relevance in drying
Huggins model based on the Hansen solubility parameters. Fluid Phase of sugar-rich foods: A review. Int J Food Prop 3(3):323–351.
Equlibria 203:247–260. 103. Benmore CJ, Weber JK, Tailor AN, Cherry BR, Yarger JL, Mou
87. Forster A, Hempenstall J, Tucker I, Rades T. 2001. Selection of Q, Weber W, Neuefeind J, Byrn SR. 2013. Structural characterization
excipients for melt extrusion with two poorly water-soluble drugs by and aging of glassy pharmaceuticals made using acoustic levitation. J
solubility parameter calculation and thermal analysis. Int J Pharm Pharm Sci 102(4):1290–1300.
226(1–2):147–161. 104. Benmore CJ, Weber JKR. 2011. Amorphization of molecular liq-
88. Meng F, Trivino A, Prasad D, Chauhan H. 2015. Investigation and uids of pharmaceutical drugs by acoustic levitation. Phys Rev X 1(1):1–
correlation of drug polymer miscibility and molecular interactions by 7.
various approaches for the preparation of amorphous solid dispersions. 105. Whiteside PT, Zhang J, Parker AP, Madden-Smith CE, Patel N,
Eur J Pharm Sci 71:12–24. Jensen J, Sloth J, Roberts CJ. 2013. Physical and chemical comparison
89. Vandecruys R, Peeters J, Verreck G, Brewster ME. 2007. Use of of material from a conventional spray-dried system and a single particle
a screening method to determine excipients which optimize the ex- spray-dried system. Int J Pharm 455(1–2):306–311.
tent and stability of super-saturated drug solutions and application 106. Jang DJ, Sim T, Oh E. 2013. Formulation and optimization of
of this system to solid formulation design. Int J Pharm 342(1–2): spray dried amlodipine solid dispersion for enhanced absorption. Drug
168–175. Dev Ind Pharm 39(7):1133–1141.
90. Warren DB, Benameur H, Porter CJH, Pouton CW. 2010. Using 107. Arpagaus C. 2011. Nano spray dryer B-90: Literature review and
polymeric precipitation inhibitors to improve the absorption of poorly applications. Accessed, at: http://www.scribd.com/doc/201979491/Nano-
water-soluble drugs: A mechanistic basis for utility. J Drug Target Spray-Dryer-B-90-Literature-Review-and-Applications on January 10,
18(10):704–731. 2015.
91. Van Eerdenbrugh B, Raina S, Hsieh YL, Augustijns P, Taylor LS. 108. Schmid K, Arpagaus C, Friess W. 2009. Evaluation of a vibrating
2014. Classification of the crystallization behavior of amorphous ac- mesh spray dryer for preparation of submicron particles. RDD Europe
tive pharmaceutical ingredients in aqueous environments. Pharm Res 2:323–326.
31(4):969–982. 109. Schmid K, Arpagaus C, Friess W. 2011. Evaluation of the nano
92. Konno H, Taylor LS. 2008. Ability of different polymers to inhibit spray dryer B-90 for pharmaceutical applications. Pharm Dev Technol
the crystallization of amorphous felodipine in the presence of moisture. 16(4):287–294.
Pharm Res 25(4):969–978. 110. Li X, Anton N, Arpagaus C, Belleteix F, Vandamme TF. 2010.
93. Konno H, Handa T, Alonzo DE, Taylor LS. 2008. Effect of polymer Nanoparticles by spray drying using innovative new technology: The
type on the dissolution profile of amorphous solid dispersions contain- BUCHI Nano Spray Dryer B-90. J Control Release 147(2):304–310.
ing felodipine. Eur J Pharm Biopharm 70(2):493–499. 111. Van den Mooter G. 2012. The use of amorphous solid dispersions:
94. Raina SA, Alonzo DE, Zhang GG, Gao Y, Taylor LS. 2014. Im- A formulation strategy to overcome poor solubility and dissolution rate.
pact of polymers on the crystallization and phase transition kinetics of Drug Discov Today Technol 9(2):e79–e85.
amorphous nifedipine during dissolution in aqueous media. Mol Pharm 112. Qian F, Wang J, Hartley R, Tao J, Haddadin R, Mathias N,
11(10):3565–3576. Hussain M. 2012. Solution behavior of PVP-VA and HPMC-AS-based
95. Meng Z, Ming Z, Wei S, Bi X, Hatch GM, Gu J, Li C. 2014. amorphous solid dispersions and their bioavailability implications.
Amorphous solid dispersion of berberine with absorption enhancer Pharm Res 29(10):2765–2776.

DOI 10.1002/jps.24541 He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015


3258 REVIEW

113. Alonzo DE, Gao Y, Zhou D, Mo H, Zhang GG, Taylor LS. 2011. 128. Alonzo DE, Zhang GZ, Zhou D, Gao Y, Taylor LS. 2009. Under-
Dissolution and precipitation behavior of amorphous solid dispersions. standing the behavior of amorphous pharmaceutical systems during
J Pharm Sci 100(8):3316–3331. dissolution. Pharm Res 27(4):608–618.
114. Augustijins P, Brewster ME. 2012. Supersaturating drug delivery 129. Hugo M, Kunath K, Dressman J. 2013. Selection of excipient,
systems: Fast is not necessarily good enough. J Pharm Sci 101(1):7–9. solvent and packaging to optimize the performance of spray-dried
115. Kararli TT. 1995. Comparison of the gastrointestinal anatomy, formulations: Case example fenofibrate. Drug Dev Ind Pharm 39(2):
physiology, and biochemistry of humans and commonly used laboratory 402–412.
animals. Biopharm Drug Dispos 16(5):351–380. 130. Harman A. 2010. A roller coaster chase for a cure. The New
116. Lust A, Laidmäe I, Palo M, Meos A, Aaltonen J, Veski P, York Times. Accessed, at: http://www.pharmacircle.com on May 10,
Heinämäki J, Kogermann K. 2013. Solid-state dependent dissolution 2015.
R
and oral bioavailability of piroxicam in rats. Eur J Pharm Sci 48(1– 131. Zelboraf (vemurafenib) tablet package insert. Accessed, at:
2):47–54. http://www.gene.com/download/pdf/zelboraf prescribing.pdf on May 10,
117. Chuah AM, Jacob B, Jie Z, Ramesh S, Mandal S, Puthan JK, 2015.
Deshpande P, Vaidyanathan VV, Gelling, RW, Patel G, Das T, Shree- 132. Miller DA, Brough CE, Keen JM. 2014. Improved formulations of
ram S. 2014. Enhanced bioavailability and bioefficacy of an amorphous vemurafenib and methods of making the same. Patent US. Provisional
solid dispersion of curcumin. Food Chem 156:227–233. patent application no. 62/074,465.
118. Forster R, Bode G, Ellegaard L, van der Laan JW. 2010. The 133. Song TT, Yuan XB, Sun AP, Wang H, Kang CS, Ren Y, He B,
RETHINK project on minipigs in the toxicity testing of new medicines Sheng J, Pu PY. 2009. Preparation of injectable paclitaxel sustained
and chemicals: Conclusions and recommendations. J Pharmacol Toxicol release microspheres by spray drying for inhibition of glioma in vitro.
Methods 62:236–242. J Appl Polym Sci 115(3):1534–1539.
119. Six K, Daems T, de Hoon J, Van Hecken A, Depre M, Bouche MP, 134. Meeus J, Chen X, Scurr DJ, Ciarnelli V, Amssoms K, Roberts CJ,
Prinsen P, Verreck G, Peeters J, Brewster ME, Van den Mooter G. 2005. Davies MC, van Den Mooter G. 2012. Nanoscale surface characteriza-
Clinical study of solid dispersions of itraconazole prepared by hot-stage tion and miscibility study of a spray-dried injectable polymeric matrix
extrusion. Eur J Pharm Sci 24(2–3):179–186. consisting of poly(lactic-co-glycolic acid) and polyvinylpyrrolidone. J
120. Mahmah O, Tabbakh R, Kelly A, Paradkar A. 2014. A comparative Pharm Sci 101(9):3473–3485.
study of the effect of spray drying and hot-melt extrusion on the prop- 135. Shiny J, Ramchander T, Goverdhan P, Habibuddin M, Aukunuru
erties of amorphous solid dispersions containing felodipine. J Pharm JV. 2013. Development and evaluation of a novel biodegradable sus-
Pharmacol 66(2):275–284. tained release microsphere formulation of paclitaxel intended to treat
121. Chiang PC, Cui Y, Ran Y, Lubach J, Chou KJ, Bao L, Jia W, La breast cancer. Int J Pharm Investig 3(3):119–125.
H, Hau J, Sambrone A, Qin A, Deng Y, Wong H. 2013. In vitro and in 136. Abraxane R
for injectable suspension package insert. Accessed, at:
vivo evaluation of amorphous solid dispersions generated by different http://www.abraxane.com/wp-content/pi/prescribing-info.html on May
bench-scale processes, using griseofulvin as a model compound. AAPS 10, 2015.
J 15(2):608–617. 137. Desai NP, Soon-Shiong P. 2000. Paclitaxel-containing formula-
122. Miller DA, DiNunzio JC, Yang W, McGinity JW, Williams III RO. tions. Patent US6753006 B1.
2008. Enhanced in vivo absorption of itraconazole via stabili-zation of 138. PharmaCircleTM database. Accessed, at: http://www.
supersaturation following acidic-to-neutral pH transition. Drug Dev pharmacircle.com on May 10, 2015.
Ind Pharm 34(8):890–902. 139. Chen Y, Liu C, Chen Z, Su C, Mageman M, Hussain M, Haskell
123. DiNunzio JC, Miller DA, Yang W, McGinity JW, Williams RO III. R, Stefanski K, Qian F. 2015. Drug-polymer-water interaction and its
2008. Amorphous compositions using concentration enhancing poly- implication for the dissolution performance of amorphous solid disper-
mers for improved bioavailability of itraconazole. Mol Pharm 5(6):968– sions. Mol Pharm 12(2):576–589.
980. 140. Yalkowsky SH. 1999. Solubility and solubilization in aqueous me-
124. Shah N, Iyer RM, Mair HJ, Choi DS, Tian H, Diodone R, Fähnrich dia. New York, NY: Oxford University Press, Inc.
K, Pabst-Ravot A, Tang K, Scheubel E, Grippo JF, Moreira SA, Go Z, 141. Yalkowsky SH, He Y, Jain P. 2009. Handbook of aqueous solubility
Mouskountakis J, Louie T, Ibrahim PN, Sandhu H, Rubia L, Chokshi data. 2nd ed. Boca Raton, FL: CRC Press.
H, Singhal D, Malick W. 2013. Improved human bioavailability of ve- 142. Kennedy M, Hu J, Gao P, Li L, Ali-Reynolds A, Chal B, Gupta V,
murafenib, a practically insoluble drug, using an amorphous polymer- Ma C, Mahajan N, Akrami A, Surapaneni S. 2008. Enhanced bioavail-
stabilized solid dispersion prepared by a solvent-controlled coprecipi- ability of a poorly soluble VR1 antagonist using an amorphous solid
tation process. J Pharm Sci 102(3):967–981. dispersion approach: A case study. Mol Pharm 5(6):981–993.
125. Ta J, Sun Y, Zhang GGZ, Yu L. 2009. Solubility of small molecule 143. Ikegami K, Tagawa K, Narisawa S, Osawa T. 2003. Suitability of
crystals in polymers: D-mannitol in PVP, in-domethacin in PVP/VA, the cynomolgus monkey as an animal model for drug absorption studies
and nifedipine in PVP/VA. Pharm Res 26(4):855–864. of oral dosage forms from the viewpoint of gastrointestinal physiology.
126. Paudel A, Van Humbeeck J, Van Den Mooter G. 2010. Theoretical Biol Pharm Bull 26(10):1442–1447.
and experimental investigation on the solid solubility and miscibility 144. Willmann S, Edginton AN, Dressman JB. 2007. Development and
of naproxen in poly(vinylpyrrolidone). Mol Pharm 7(4):1133–1148. validation of a physiology-based model for the prediction of oral absorp-
127. Qian F, Huang J, Zhu Q, Haddadin R, Gawel J, Garmise R, tion in monkeys. Pharm Res 24(7):1275–1282.
Hussain M. 2010. Is a distinctive single Tg a reliable indicator 145. Miller DA, DiNunzio JC, Yang W, McGinity JW, Williams III RO.
for the homogeneity of amorphous solid dispersion? Int J Pharm 2008. Targeted intestinal delivery of supersaturated itraconazole for
395(1–2):232–235. improved oral absorption. Pharm Res 25(6):1450–1459.

He and Ho, JOURNAL OF PHARMACEUTICAL SCIENCES 104:3237–3258, 2015 DOI 10.1002/jps.24541

You might also like