Vol 2, Issue 2, 2020 (55-68)
http://journal.unpad.ac.id/idjp
*Corresponding author,
e-mail : maria18015@mail.unpad.ac.id (M. E. T Butarbutar)
https://doi.org/10.24198/idjp.v2i2.27123
2020 M. E. T Butarbutar et al
Characterization methods of amorphous form stability in solid dispersion: A review
Maria Elvina Tresia Butarbutar
1,*
, Nasrul Wathoni
2
, Yoga Windhu Wardhana
1
1
Magister Program of Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363,
Indonesia.
2
Departement of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Jatinangor 45363
Received : 2 May 2020/Revised : 12 May 2020/Accepted : 17 May 2020/ Published:23 Jun 2020
ABSTRACT
Solubility as a cause of ineffective active pharmaceutical ingredients (API) needs to be a
concern. One of the solutions to increase the solubility by choosing active ingredients in the
amorphous form. However, the amorphous form tends to be unstable because it has high Gibbs
free energy and molecular mobility. To overcome those properties solid dispersion methods can
be an answer. The dispersion of the amorphous form in the polymer is expected to prevent the
transformation of API to crystal stable form. The solid dispersion (SD) resulted needs for
physicochemical characterization to prove the ability of SD to maintain the amorphous form.
Therefore, the physicochemical properties of the amorphous solid dispersions (ASDs) have to
analyze there in any interactions that are able to occur between the drug and the polymer. Also
for evaluate the stability of the ASDs within a certain period. In the article presents, some
articles related with ASDs and its characterization will studying, include several product on the
market as example. The number of literature used in this article is 69 articles.
Keywords: Solubility, amorphous for, solid dispersion, characterization ASDs.
Abbreviations
DSC Differential scanning calorimetry
FTIR Fourier transform infrared
HSPLM Hot stage polarization light microscopy
PLM Polarization light microscopy
SEM Scanning electron microscopy
ssNMR Solid state nuclear magnetic resonance
TGA Thermogravimetric analysis
TMDSC Temperature modulated differential scanning calorimetry
1. Introduction
More than 75% of commercial pharmaceutical
oral solid dosage forms have poor water solubility
(1). This properties will affect the availability of
biological and therapeutic activities. Several factors
that affect solubility of the API which,
polymorphism (2), crystallinity (3), particle size (4),
pKa (1), dielectric constant, and polarity (5). One
effort to improve API solubility is choosing an
amorphous forms which has better solubility than a
stable crystalline form (3). This amorphous form
properties come from lacks lattice energy, random
molecular structure, and large surface area. Besides
that, the amorphous form is unstable and tends to
M. E. T Butarbutar et al / Indo J Pharm 1 (2020) 55-68
56
undergo a molecular transformation in its crystalline
stable form. The instability occurs due to high Gibbs
free energy and large molecular mobility (6). The
strengths and weaknesses of the amorphous form
characterize become a solution and also a challenge
for the pharmaceutical development.
To overcome this challenge there are methods
that are widely used to improve amorphous stability.
Solid dispersion is one of the methods, with several
techniques, including fusion methods, solvent
methods, hot melt extraction, spray drying, freeze
drying, supercritical fluid, electrostatic spinning, and
lyophillization (6). In solid dispersion methods
polymer is generally use as a matrix. The polymer
has capable inhibit the bonds orientation between
intramolecules and to detain nucleation. In addition,
the hydrophilic polymer is also able to improve
wettability (7). Characteristics of polymer that can
be used as matrix, among others thermal stability at
melting temperature, melting point not much higher
than that of drug, low vapour pressure, hight
molecule weight, and thet should be non toxic (8).
In 2007 to 2017 the Food and Drug and
Administration (FDA) has been commercially
allowed amorphous solid dispersion (ASD) products
(9). Several examples of commercial ASDs
products, namely Novir
®
with Ritonavir as API and
using polyvinylpyrolidone (PVP) as a polymer (10).
Heterogeneous ASDs are give rise to interactions
between drug molecules and polymers that the alter
kinetic stability of the drug. In this article presents
several aspects regarding ASDs, namely amorphous
instability, schematic conversion of API crystalline
form from amorphous in solid dispersion systems,
characterization of ASDs, and example of ASDs
product that have been circulating in the market.
2. Methodology
This review is based on the literature obtained
from the at Google Scholar, Sciendirect database
2010 2019 using the keywords “Amorphous solid
dispersion” and “Solid dispersion” and
“Characterization amorphous solid dispersion” and
“Amorphous form and “Development of solid
dispersion” and “evaluation of solid dispersion”.
Figure 1. Flow chart of the reference search
3. Discussion
3.1. Amorphous solid dispersion
Solid dispersions embed active pharmaceutical
ingredients (APIs) in polymeric carriers to improve
their solubility. Generally, in solid dispersion
systems, a highly hydrophilic polymer is used to
increase the solubility of API, high T
g
than Tg API to
maintain API stability of API molecules, high
fragility, and hydrogen donors / acceptors because
can helped inhibit crystallization. In addition, the
thermal characteristics and molecular weight of a
polymer will affect the effectiveness of the polymer
as a stabilizer (11). The properties of the polymer
stabilizer in ASDs are also due to the increase in T
g
which causes decreased molecular mobility and
increased stability (7). Implement this mechanism
have shown the success of increased solubility and
stability of the drug in such established products
(1214).
Initial selected article (n = 97)
69 articles selected after initial screening
28 articles excluded
Years article <
2010
Vol 2, Issue 2, 2020 (55-68)
http://journal.unpad.ac.id/idjp
*Corresponding author,
e-mail : maria18015@mail.unpad.ac.id (M. E. T Butarbutar)
https://doi.org/10.24198/idjp.v2i2.27123
2020 M. E. T Butarbutar et al
Figure 2. Schematic crystalline drug which becomes an ASDs and is characterized (15)
As demonstrated in Figure 2, molecular
interactions occur due the mixing of API and
polymer in a completely soluble state. A soluble API
will form smaller particles that immediately
suspended into a polymer solution producing a
heterogeneous (16). The preparation based on the
characteristic of API material. Well known
characteristic of API are essential to achieve
successful ASDs. For example, the spray drying
preparation is used for API that are soluble in
volatile solvents (17), a co-precipitation method is
used for API with high melting points and low
solubility in organic solvents. For API with high
melting points, the hot melt extrusion is used instead
(18), solvent evaporation method is used for drugs
that are unstable to heating drug and carrier are
mixed by a solvent instead of heat as in melting
method (19). Polymer applications and solid
dispersion preparation methods are presented in
Table 1.
Based on Table 1, the hot melt extrusion
preparation is the most widely used method. The
absence of organic solvent yields a result without
residue. This method involves mixing, melting, and
homogenizing which can be applied on an industrial
scale. For example, oleanolic acid prepared using hot
melt extrusion using PVP VA64, shows a dissolution
rate of 90% in 10 minutes (18). Also, efavirenz
which was also prepared using hot melt extrusion
using Plasdone S-630 showed an increase in
dissolution rate of 1.7 fold (12).
Because ASDs are molecular, characterization
methods are needed to investigate the amorphous
form. Several methods are required to complete the
amorphous form information in solid dispersions.
Characterization methods that can be performed
such as thermal analysis (DSC, TMDSC, TGA) to
analyze changes in the thermal properties of
components, vibrational spectroscopy (FTIR,
Raman) to analyze the interaction of component
molecules, and XRD to analyze changes in
crystallinity as presented in Table 2. The
characterization method used in ASDs. Each
characterization method will support each other to
aid the investigation of amorphous forms in solid
dispersions. The next section will discuss the
interpretation of result of characterization.
3.2 Characterization of Amorphous by Solid
Dispersion and Applications
3.2.1 Spectroscopic Vibrational
Spectroscopy is a tool often used to investigate
interactions between drugs and polymers in solid
Solvent base
Rapid cooling (quenching) of a melt
Solvent free
Crystal drug
Polymer
Amorphous
solid dispersion
Spectroscopy
Microscopy
Thermal analysis
Morphological
Stability
Application ASDs
M. E. T Butarbutar et al / Indo J Pharm 1 (2020) 55-68
58
Table 1. Example of different polymers and technique used in the formulation ASDs
API
BCS
class
Polymers / Carrier
Preparations
method
Year
Ref
Amphotericin B
IV
Poly-cyclodextrin
Spray drying
2018
(20)
Baicalin
Hydroxypropyl-β-
Cyclodextrin
Supercritical fluid
encapsulation
2018
(21)
Carvedilol
II
Eudragit
®
, Soluplus
®
,
HPMCAS
Supercritical CO
2019
(22)
Efavirenz
II
Eudragit EPO, Plasdone S-630
Hot Melt Extrusion
2012
(12)
Felodipine
II
Soluplus
®
Hot Melt Extrusion
2016
(23)
Griseofulvin
II
Kollidone
®
VA64
Melt-quench
method
2019
(24)
Hydrocortison
II
PEG 4000, Kolliphor
®
P-407
Spray drying
2019
(25)
Ibuprofen
II
HPMCAS, HPMCP HP-55
Electrospinning
2019
(26)
Nilotinib
II
Soluplus
®
Spray dried
2017
(14)
Nifedipine
II
Kollidon
®
VA64, Kollidon
®
12 PF, Kollidon
®
25,
Kollidon
®
90F
Hot Melt Extrusion
2019
(15)
Oleanolid acid
IV
PVP VA64
Hot Melt Extrusion
2017
(13)
Pyrimethamine
III
PVP K-25, PEG 6000,
Poloxamer 188
Solvent evaporation
2018
(27)
Ritonavir
IV
PVPVA64
Solvent evaporation
2019
(28)
Zoplicone
II
PVP K-25
Solvent evaporation
/ Freeze drying
2015
(29)
dispersion systems. Changes that occur, such as
vibrations, homomolecular, and heteromolecular
transitions. This method involves electromagnetic
waves that interact with materials and cause a
transition between vibrations with the energy of the
molecule. Spectroscopy can provide information
from macro, micro, and nano scales (30). The data
gathered in the form of a spectrum that describes the
intensity of transmission.
FTIR and Raman
FTIR (4000 400 cm
-1
) and Raman (4000 150
cm
-1
) are very efficient techniques among the
methods of vibration spectroscopy. FTIR can
analyze ASDs, which correlate with molecular
structure and donor-acceptor characteristics that
form hydrogen bonds between drugs and polymers
and establish miscibility (31). In addition, FTIR can
also conclude the formation of ASDs. ASDs
ibuprofen-HPMCAS (1:9) products peak expansion
which signifies amorphous presence of ibuprofen at
wavelengths 1231 cm
-1
and 1721 cm
-1
. In the
spectrum that describes the amorphous form,
wavelength stretching occurs at 1231 cm
-1
and 1721
cm
-1
which signifies a shift in the carboxyl group,
illustrating the presence of hydrogen bonds between
carboxylic acids from ibuprofen and hydroxyl
groups from HPMCAS (26). Raman on the other
hand, is an innovative molecular analysis to evaluate
ASDs. The Raman is more sensitive and has a wider
spectrum. The spectrum difference between ASDs
and crystalline nitredipin can be seen clearly on
Raman. Li et al. explains the significant difference in
wavelength intensity between nitredipine and
nitredipine-PVP crystals. Nitredipine crystals show
sharper wavelength than ASDs products. This is
supported by the shifting of the C-O-C band at
wavelengths of 1680 and 1700 cm
-1
which indicates
the formation of hydrogen bonds from nitrendipine-
PVP and confirms the amorphous state (32). These
results showed that both FTIR and Raman provided
clear information in analyzing ASDs.
M. E. T Butarbutar et al / Indo J Pharm 1 (2020) 55-68
59
ssNMR
ssNMR is a non-destructive technique, providing
information both qualitatively and quantitatively, it
can also be 1-dimensional or 2-dimensional with
details based on spectroscopic techniques,
relaxometry, and structural images (33). Its main
purpose is to identify the structure and
transformation of solids. The prediction made by
ssNMR regarding the domain size between the drug
and the polymer by correlating the relaxation time
with the round diffusion length scale (34).
Foster et al., comparing Nifedipine and
indomethacin ASDs mobility using
1
H NMR. The
relaxation behaviour of nifedipine ASDs
significantly in comparison compared to
indomethacin ASDs that explain that the stability of
ASDs nifedipine is lower. However, molecular
mobility is not the only parameter to predict
nucleation and the rate of crystal growth (35). Skiba
et al. uses
13
C NMR against cyclosporine ASDs
αβ-Cyclodextrins, cyclosporine crystal spectrum
with alkyl C-C peak chemical intensity between 30
and 10, N-C = O at 174 170; C = O at 130 120;
C-OH at 75 70; and N at 60 50 ppm were
changed after being on a solid dispersion system. C-
C alkyl intensity significantly changes with the
occurrence of chemical shifts. It signifies the
interaction of cyclosporine-αβ-cyclodextrins
converted into an amorphous form (36). Note that
the mixing method also affects ssNMR spectrum
results.
3.2.2 Microscopic and Morphological
Microscopes are widely used for characterization
of solids. Information obtained includes
morphology, particle size and crystallinity of the
sample (37). Characterization of ASDs generally
uses polarized light microscopy (PLM) because it
can detect crystals or amorphous. Amorphous solids
are isotropic, which means the molecules are
randomly oriented. As a result, amorphous solids do
not have double refraction, are nonbirefringent, and
do not show color interference. Meanwhile,
crystalline solids are anisoptric whose molecules are
arranged in a regular order, have color interference
to detect based on the birefringence. In addition, hot
stage polarized microscopy (HSPLM) is also widely
used in the pharmaceutical field, except that HSPLM
can observe the thermal behavior of a sample. The
heating and cooling rates can be controlled with
accurate results (38).
PLM and HSPLM
The PLM application was carried out by Kim et
al. by the lightly grinding method, which is between
dutasteride-polymer (Kollicoat
®
MAE 100P,
Soluplus
®
, Lutrol
®
F68, and Kollidon
®
K30.
Microscopic observations were made to see the
solubility of the dutasteride dispersed in a polymer
(Fig. 3). It was assumed that the higher the sample
clarity from the PLM results then it also had the
higher solubility capacity. Based on PLM results,
dutasteride - Kollicoat
®
MAE 100P had a higher
solubility (39).
Figure 3. Polarized microscopic view of dutasteride
solid dispersion (39)
Parikh et al. use PLM to detect the presence of
itraconazole crystals dispersed in Soluplus
®
. The
resulting polarization images show the presence of
microcrystals birefringence that was not previously
detected by DSC and XRD. It should be noted that
the presence of microcrystals is also influenced by
the ratio between API-polymers. Based on this, PLM
is a more sensitive method compared to XRD and
DSC for analyzing crystal growth (54).
M. E. T Butarbutar et al / Indo J Pharm 1 (2020) 55-68
60
Table 2. Solid-state analytical techniques used to study ASDs
API / Sample
Processing method
(s)
Solid state analytical technique (s)
Spectroscopy
Microscopy
X-Ray
Thermal and
Gravimetric
Others
FTIR
Raman
UV-Vis
ssNMR
MRI
SEM
HSM
PLM
OM
XRD
DSC
TMDSC
DTA
TGA
PSA
HPLC
PSD
Year of
publication
Ref
Aprepitant
Spray drying
2016
(40)
Amphotericin B
Spray drying
2018
(20)
Aripiprazole
Hot Melt Extrusion
2019
(41)
Atorvastatin
Solvent evaporation
2018
(42)
Betulinic acid
Gently grinding
2015
(43)
Bicalutamide
Fusion method
2015
(44)
Carbamazepine
Hot Melt Extrusion
2015
(45)
Carvedilol
Supercritical CO
2019
(22)
Cyclosporine
Spray drying
2018
(36)
Darunavir
Electrospraying
2018
(46)
Griseofulvin
Melt-quench method
2019
(24)
Hydrocortisone
Spray drying
2019
(25)
Itraconazole
Hot Melt Extrusion
2017
(47)
Lapatinib
Solvent evaporation
2018
(48)
Lopinavir
Solvent evaporation
2018
(49)
Leflunomide
Freeze drying
2017
(50)
Nifedipine
Hot Melt Extrusion
2019
(15)
Nimodipine
Hot Melt Extrusion
2018
(51)
Nitrendipine
Solvent evaporation
2017
(31)
Pyrimethamine
Solvent evaporation
2018
(27)
Ritonavir
Solvent evaporation
2019
(52)
Tadalafil
Spray drying
2015
(53)
M. E. T Butarbutar et al / Indo J Pharm 1 (2020) 55-68
61
Unlike HSPLM which can visually determine the
melting and thermal transition levels. The HSPLM
application is used against the ASDs nifedipine-PVP
which demonstrates the visualization differences
between ASDs nifedipine and crystalline nifedipine
prior to melting, the onset of melting, when melted,
and after melting perfectly. HSPLM provides
information that the transformation of crystal form
into amorphous form (15).
SEM
SEM has been widely used in the pharmaceutical
field to develop and control the quality of materials
and dose. The main purpose of using SEM is to
analyze the morphology of the sample, particle size,
and formulation so that it can be used to analyze
ASDs. Quantitative analysis can also be obtained
(55). The electrons in the SEM interact with the
sample resulting in a complex signaling mechanism.
Specimen interaction with electrons can be classified
into two groups, i.e. elastic and non-elastic collision.
Elastic collision occurs when electrons strike the
specimen and change the direction without losing
energy. Whereas non-elastic collision does not
change direction, but transfers the electrons to the
specimen (56).
Das et al. used SEM to characterize the
morphology of pure ibuprofen and ASDs ibuprofen-
captisol
®
(Fig. 4)
.
The morphology of ibuprofen was
shown as rectangular crystals with a smooth surface
while the morphology of captisol
®
was spherical.
The morphology of ASDs ibuprofen changed with
the loss of rectangular shape (57).
Figure 4. Scanning electron micrograph (a) Ibuprofen, (b) Captisol, (c) Ibuprofen-Captisol
®
(58)
The use of SEM was also carried out by Ha et al.
to analyze celecoxib-PVP K30 ASDs. The used
nano-size particles were dispersible in PVP K30.
ASDs celecoxib-PVP K30 were spherical
nanoparticles with particle sizes ranging from 150
158 nm and surface area from 78 81 m
2
/g. SEM
results showed that the morphology of ASDs
celecoxib did not differ significantly from pure
celecoxib. However, particle size and surface area
differed significantly. This was influenced by the
addition of surfactants (59).
3.2.3 X-Ray Diffraction
XRD is an analytical method that can show
specific molecular materials such as crystals,
mesophases, amorphous, and mixtures. Not only in
the form of powder, now XRD can also analyze
intact tablet preparations (60). The appearance of the
diffraction peak indicates the crystalline molecule,
whereas the absence of the diffraction peak indicates
that the molecule is amorphous (61). The
information gathered is in the form of a
diffractogram consisting of intensity and an angle of
2. XRD application is widely used to study the
kinetics of crystallization in solid dispersion
systems. Many factors also influence the
diffractogram, including the temperature of the
treatment and the method of manufacture. Examples
of applications using XRD, the kinetics of
crystallization of ASDs naproxen-polyethylene
glycol reduced compared to pure naproxen which
was affected by temperature differences. At the
temperature of 25 °C, naproxen crystallizes faster
M. E. T Butarbutar et al / Indo J Pharm 1 (2020) 55-68
62
than at 40 °C. However, the addition of polyethylene
glycol can slow the crystallization that happened at
40 °C. Additionally the addition of polyethylene
glycol results in a smaller naproxen domain in the
matrix. In addition, other studies conducted by
Surana et al. compared crystallization kinetics based
on amorphous manufacturing methods, i.e. the
method of melt quenching, spray drying, freeze
drying, and dehydration of trehalose dihydrate. The
results of the study found that the dehydration of
trehalose dihydrate had the fastest crystallization.
This was caused by differences in molecular nucleic.
In this study it can be concluded that there are
significant differences from each method on the
results of the XRD analysis (62).
Homayouni et al. also performed celecoxib-PVP-
K30 ASDs analysis. Pure celecoxib diffraction
showed the formation of the crystal presented by the
sharp peaks at a value of at 5°; 10.5°; 16°; and
21.5° (Fig. 5).
Figure 5. X-ray powder diffractions of pue
celecoxib (CLX), polyvinylpyrrolidone K-30 (PVP)
and CLX : PVP K-30 (63).
Whereas, PVP-K30 showed amorphous nature
due to the absence of diffraction peaks. In a solid
dispersion system using the solvent evaporation
method, ASDs celecoxib-PVP-K30 did not find
diffraction peaks. This indicates that celecoxib
crystals have been converted to amorphous in a solid
dispersion system (61).
To this date, XRD is one of the most popular
methods of analyzing ASDs because it provides very
informative results regarding the conversion of
crystals to amorphous, and vice versa.
3.2.4 Thermal Analysis
Thermal analysis is used to study the properties of
materials affected by temperature. Information that
can be obtained, such as glass transition (T
g
),
melting temperature (T
m
), decomposition
temperature (T
d
), crystallization temperature (T
c
),
transformation polymorphism, crystallization,
desolvation, entalphi value, and changes in chemical
reactions in the form of endothermic reactions (heat
absorption) or exothermic (heat release) (64). The
occurrence of T
m
indicates the characteristics of a
solid that is changing enthalpy, such as crystal
solids, whereas T
g
indicated amorphous because it is
considered as a point of transition (65). Looking at
ASDs consisting of complex components (API and
polymer) allows interactions that affect the
thermodynamic nature of the drug. The methods
included in thermal analysis are DSC, TMDSC,
DTA, and TGA, information obtained is in the form
of a thermo gram that describes the thermal
characteristics of the sample being analyzed.
DSC and TMDSC
DSC and TMDSC methods that can analyze up to
the molecular level. However, DSC has a
disadvantage, that is, thermogram interpretation can
be hindered if two or more thermal events overlap.
Unlike the case with TMDSC, fast-scans on very
sensitive TMDSC can separate complex overlapping
thermal events. TMDSC can also detect glass
transition temperatures and amorphous phase
quantification accurately, and the availability of
relaxation time calculations when phase separation
occurs. But will, the downside of these thermal
analysis results is that it cannot accurately analyze
the events that occur concurrently and will result in
overlap (56).
In ASDs, drug mixtures with polymers must be
soluble and homogeneous because they affect the
thermodynamic profile. It is expected that ASDs
have high T
g
and low enthalpy so that they are
M. E. T Butarbutar et al / Indo J Pharm 1 (2020) 55-68
63
physically stable. Wlodarski et al. to see the
thermodynamic properties of ASDs tadalafil-
PVPVA using spray drying method. DSC provided
information on T
g
of crystalline tadalafil on 149.9°C
followed by an exothermic reaction with an onset of
181.6 °C and Tm of endothermic reaction is shown
at peak on 302.1 °C (Fig. 6).
Figure 6. DSC thermograms (from top to bottom)
crystalline tadalafil (Td), spray dried tadalafil, 9:1
1:9 (w/w) Td-PVPVA solid dispersion (66).
In a solid dispersion system, the peak
endothermic reaction experienced a change in T
m
under the endothermic reaction of crystalline
tadalafil amorphous molecular enthalpy relaxation
usually occurs around T
g
(67).
In its application the concentration of API:
polymer contributes to T
g
, such as T
g
betulinic acid
188.81 ° C, T
g
74.25. In solid dispersion systems
with a ratio of 1:4 T
g
ASDs to 77.45 ° C. This T
g
value also affects the solubility of betulinic acid
ASDs which increases 15-fold (43). From this
example it can be concluded that the polymer can
increase T
g
of API and slow down the mobility of
amorphous molecules as well as inhibit the
crystallization process.
TGA
TGA is generally combined with other methods,
such as DSC and FTIR. In the characterization of
ASDs, TGA is used to determine thermal stability,
changes in material weight affected by temperature,
decomposition, the profile of volatile components. It
is important to note the thermal characteristics of
each component to determine the temperature range
used in TGA applications. Research conducted by
Mehenni et al., Using TGA with a temperature range
of 30 - 150 ° C. amphotericin B 3.40%, poly γ-CD
5.69%, while the dispersion system experienced an
weight loss of 1.87%, which is the percentage of
evaporation of water that depends on relative
humidity. This percentage decrease indicates that
there is an interaction between amphotericin B and
poly γ-CD (20). Aripiprazole ASDs prepared by the
hot melt extrusion method using PVP experienced a
weight loss of 5% (Fig. 7).
Figure 7. Thermogravimetric curves of Aripiprazole
(ARP), succinic acid (SA), physical mixture (PM),
and polyvinylpyrrolidone (PVP) (41).
Because PVP is highly hydrophilic which
contains about 5% water, when affected by
temperatures of 60 - 220 °C the water is evaporated
(41). In terms of the results obtained, the TGA does
not provide specific information regarding the state
of the amorphous form in solid dispersions.
3.3 Products of Amorphous Solid Dispersion
Amorphous in the solid dispersion system has
demonstrated its success in overcoming the
solubility of poorly water soluble drugs and the
method of solid dispersion in maintaining
amorphous stability. Therefore, the FDA has
approved several ASDs products that have been
circulating in the market, among which are presented
in Table 3.
M. E. T Butarbutar et al / Indo J Pharm 1 (2020) 55-68
64
Table 3. Amorphous solid dispersion products on the market (9,68)
API
BCS
Indication
Polymer
Preparation
method
Product
Year of
acc.
Nabilone
IV
Antiemetic
PVP
-
Cesamet
1985
Itroconazole
II
Antifungal
HPMC
Spray drying
Sporanox
®
1992
Tacrolimus
II
immunosuppressive
HPMC
Spray drying
Prograf ™
1994
Griseofluvin
II
Antigungal
PEG
Melt extruction
Gris-PEG
2000
Lopinavir
IV
Antiretroviral
PVP-VA
Melt extruction
Kaletra
®
2005
Etravirine
IV
Antiretroviral
HPMC
Spray drying
Intelence
®
2008
Ritonavir
IV
Antiretroviral
PVP-PA
Melt extruction
Novir
®
2010
Ivacaftor
II
Cystic fibrosis
HPMCAS
Spray drying
Kalydeco
®
2010
Everolimus
IV
immunosuppressive
HPMC
Spray drying
Zotress
®
2010
Itroconazole
II
Antifungal
HPMC
Melt extruction
Onmel
®
2010
Fenofibrate
II
Hypercholesterolemia
PEG/Poloxamer
188
Spray melt
Fenoglide
2010
Telaprevir
II
Antiviral
HPMCAS
Spray drying
Incivek
®
2011
Vemurafenib
IV
Melanoma
HPMCAS
Coprecipitation
Zelboraf
®
2011
Posaconazole
II
Antifungal
HPMCAS
Melt extruction
Noxafil
®
2013
Ombitasvir
IV
Antiviral
PVP-VA/TPGS
Melt extruction
Viekirax
®
2014
Lumacaftor
II
Cystic fibrosis
HPMCAS
Spray drying
Orkambi
®
2015
So far there have been >15 ASDs products
circulated in the market. Among them, tacrolimus
prepared by spray drying method using HPMC
showed an increase in solubility of 25 fold, C
max
10
fold, and AUC 9.9 fold of crystal for. Ziprasiode
hydrochloride prepared by merely mixing HPMCAS
showed an increase of 1.5 fold solubility of the
crystal form. Additionaly, CAPHth / itaconazole
ASDs show a 2 fold increase in biovailability of the
crystal form (69). Many ASDs have demonstrated
success in increasing the solubility, dissolution rate,
and bioavailability of previous products.
Considering the advantages and potential offered by
ASDs with more efficient therapeutic products, it is
not that surprising there are currently many
researches focused on ASDs.
4. Conclusion
Amorphous API forms dispersed in hydrophilic
polymer have shown success in increasing solubility.
Each ASDs preparation method must take notice to
the thermal and solubility characteristics of API and
polymer. Hot melt extrusion is a preparation method
that is widely used because it does not cause residues
and has produced good quality ASDs. Because of its
molecular nature, ASDs evaluated for detecting
amorphous stability using various methods such as
PLM which is very sensitive to the presence of
microcrystals, XRD with no peaks in the
diffractogram that indicate amorphous form, and
DSC to changes in the thermal characteristic
components of ASDs. Reviewing the success of
ASDs from the increase in solubility, dissolution
rate, and bioavailability of previous products,
currently as many as >15 patent products have been
circulating in the market and become promising
products in the future.
5. Acknowledgements
Author would like to thank lecturer Dr. Yoga
Windhu Wardhana, M. Si., Apt. and Nasrul
Wathoni, Ph.D., Apt. for his guidance and support
for the drafting of this article.
M. E. T Butarbutar et al / Indo J Pharm 1 (2020) 55-68
65
References
1. Williams HD, Trevaskis NL, Charman SA,
Shanker RM, Charman WN, Pouton CW, et al.
Strategies to address low drug solubility in
discovery and development. Pharmacol Rev.
2013;65(1):315499.
2. Ku MS, Dulin W. A biopharmaceutical
classification-based Right-First-Time
formulation approach to reduce human
pharmacokinetic variability and project cycle
time from First-In-Human to clinical Proof-Of-
Concept. Pharm Dev Technol. 2012;17(3):285
302.
3. Di L, Fish P V., Mano T. Bridging solubility
between drug discovery and development. Drug
Discov Today. 2012;17(910):48695.
4. Khadka P, Ro J, Kim H, Kim I, Kim JT, Kim H,
et al. Pharmaceutical particle technologies: An
approach to improve drug solubility, dissolution
and bioavailability. Asian J Pharm Sci.
2014;9(6):30416.
5. Sachin Parmar, Amit Gangwal
NSD.Scholar.Scholars Research Library.
2011;2(4):37383.
6. Mansour RSH, Deb PK, Tekade RK. Role of
Amorphous State in Drug Delivery. Dosage
Form Design Parameters. 2018. 105154 p.
7. He Y, Ho C. Amorphous Solid Dispersions:
Utilization and Challenges in Drug Discovery
and Development. J Pharm Sci.
2015;104(10):323758.
8. Kumar DP, Dispersions S, Kumar DP, Vandana
A. Journal of Pharmaceutical and Scientific
Innovation SOLID DISPERSIONS
 : A
REVIEW. 2012;1(June):2734.
9. Jermain S V., Brough C, Williams RO.
Amorphous solid dispersions and nanocrystal
technologies for poorly water-soluble drug
delivery An update. Int J Pharm. 2018;535(1
2):37992.
10. Baghel S, Cathcart H, O’Reilly NJ. Theoretical
and experimental investigation of drug-polymer
interaction and miscibility and its impact on
drug supersaturation in aqueous medium. Eur J
Pharm Biopharm. 2016;107:1631. Available
from:
11. Mazumder S, Dewangan AK, Pavurala N.
Enhanced dissolution of poorly soluble antiviral
drugs from nanoparticles of cellulose acetate
based solid dispersion matrices. Asian J Pharm
Sci. 2017;12(6):53241.
12. Sathigari SK, Radhakrishnan VK, Davis VA,
Parsons DL, Babu RJ. Amorphous-state
characterization of efavirenz-polymer hot-melt
extrusion systems for dissolution enhancement.
J Pharm Sci. 2012;101(9):345664.
13. Gao N, Guo M, Fu Q, He Z. Application of hot
melt extrusion to enhance the dissolution and
oral bioavailability of oleanolic acid. Asian J
Pharm Sci. 2017;12(1):6672.
14. Herbrink M, Schellens JHM, Beijnen JH, Nuijen
B. Improving the solubility of nilotinib through
novel spray-dried solid dispersions. Int J Pharm.
2017;529(12):294302.
15. Ma X, Huang S, Lowinger MB, Liu X, Lu X, Su
Y, et al. Influence of mechanical and thermal
energy on nifedipine amorphous solid
dispersions prepared by hot melt extrusion:
Preparation and physical stability. Int J Pharm.
2019;561:32434.
16. Van Drooge DJ, Hinrichs WLJ, Visser MR,
Frijlink HW. Characterization of the molecular
distribution of drugs in glassy solid dispersions
at the nano-meter scale, using differential
scanning calorimetry and gravimetric water
vapour sorption techniques. Int J Pharm.
2006;310(12):2209.
17. Paudel A, Worku ZA, Meeus J, Guns S, Van
Den Mooter G. Manufacturing of solid
dispersions of poorly water soluble drugs by
spray drying: Formulation and process
considerations. Int J Pharm. 2013;453(1):253
84.
18. Shah S, Maddineni S, Lu J, Repka MA. Melt
extrusion with poorly soluble drugs. Int J
Pharm. 2013;453(1):23352.
19. Tran P, Pyo Y-C, Kim D-H, Lee S-E, Kim J-K,
Park J-S. Overview of the Manufacturing
Methods of Solid Dispersion Technology for
Improving the Solubility of Poorly Water-
Soluble Drugs and Application to Anticancer
Drugs. Pharmaceutics. 2019;11(3):132.
M. E. T Butarbutar et al / Indo J Pharm 1 (2020) 55-68
66
20. Mehenni L, Lahiani-Skiba M, Ladam G,
Hallouard F, Skiba M. Preparation and
Characterization of Spherical Amorphous Solid
Dispersion with Amphotericin B.
Pharmaceutics. 2018;10(4):235.
21. Li Y, He ZD, Zheng QE, Hu C, Lai WF.
Hydroxypropyl-β-cyclodextrin for delivery of
baicalin via inclusion complexation by
supercritical fluid encapsulation. Molecules.
2018;23(5).
22. Milovanovic S, Djuris J, Dapčević A,
Medarevic D, Ibric S, Zizovic I. Soluplus
®
,
Eudragit
®
, HPMC-AS foams and solid
dispersions for enhancement of Carvedilol
dissolution rate prepared by a supercritical CO
2
process. Vol. 76, Polymer Testing. Elsevier Ltd;
2019. 5464 p.
23. Lu J, Cuellar K, Hammer NI, Jo S, Gryczke A,
Kolter K, et al. Solid-state characterization of
Felodipine-Soluplus amorphous solid
dispersions. Drug Dev Ind Pharm.
2016;42(3):48596.
24. Zheng K, Lin Z, Capece M, Kunnath K, Chen L,
Davé RN. Effect of Particle Size and Polymer
Loading on Dissolution Behavior of Amorphous
Griseofulvin Powder. J Pharm Sci.
2019;108(1):23442.
25. Altamimi MA, Elzayat EM, Qamar W, Alshehri
SM, Sherif AY, Haq N, et al. Evaluation of the
bioavailability of hydrocortisone when prepared
as solid dispersion. Saudi Pharm J. 2019;
26. Ziaee A, O’Dea S, Howard-Hildige A, Padrela
L, Potter C, Iqbal J, et al. Amorphous solid
dispersion of ibuprofen: A comparative study on
the effect of solution based techniques. Int J
Pharm. 2019;118816.
27. Khatri P, Shah MK, Patel N, Jain S, Vora N, Lin
S. Preparation and characterization of
pyrimethamine solid dispersions and an
evaluation of the physical nature of
pyrimethamine in solid dispersions. J Drug
Deliv Sci Technol. 2018;45:11023.
28. Indulkar AS, Lou X, Zhang GGZ, Taylor LS.
Insights into the Dissolution Mechanism of
Ritonavir-Copovidone Amorphous Solid
Dispersions: Importance of Congruent Release
for Enhanced Performance. Vol. 16, Molecular
Pharmaceutics. 2019. 13271339 p.
29. Milne M, Liebenberg W, Aucamp M. The
Stabilization of Amorphous Zopiclone in an
Amorphous Solid Dispersion. AAPS
PharmSciTech. 2015;16(5):1190202.
30. Ma X, Williams RO. Characterization of
amorphous solid dispersions: An update. J Drug
Deliv Sci Technol. 2019;50(November
2018):11324.
31. Li J, Fan N, Li C, Wang J, Li S, He Z. The
tracking of interfacial interaction of amorphous
solid dispersions formed by water-soluble
polymer and nitrendipine. Appl Surf Sci.
2017;420:13644.
32. Lust A, Strachan CJ, Veski P, Aaltonen J,
Heinämäki J, Yliruusi J, et al. Amorphous solid
dispersions of piroxicam and Soluplus
®
:
Qualitative and quantitative analysis of
piroxicam recrystallization during storage. Int J
Pharm. 2015;486(12):30614.
33. Paudel A, Geppi M, Van Den Mooter G.
Structural and dynamic properties of amorphous
solid dispersions: The role of solid-state nuclear
magnetic resonance spectroscopy and
relaxometry. J Pharm Sci. 2014;103(9):2635
62.
34. Yuan X, Sperger D, Munson EJ. Investigating
miscibility and molecular mobility of
nifedipine-PVP amorphous solid dispersions
using solid-state NMR spectroscopy. Mol
Pharm. 2014;11(1):32937.
35. Tombari E, Ferrari C, Johari GP, Shanker RM.
Calorimetric relaxation in pharmaceutical
molecular glasses and its utility in
understanding their stability against
crystallization. J Phys Chem B.
2008;112(35):1080614.
36. Lahiani-Skiba M, Hallouard F, Bounoure F,
Milon N, Karrout Y, Skiba M. Enhanced
Dissolution and Oral Bioavailability of
Cyclosporine A: Microspheres Based on αβ-
Cyclodextrins Polymers. Pharmaceutics.
2018;10(4):285.
37. Guo Y, Shalaev E, Smith S. Physical stability of
pharmaceutical formulations: Solid-state
M. E. T Butarbutar et al / Indo J Pharm 1 (2020) 55-68
67
characterization of amorphous dispersions.
TrAC - Trends Anal Chem. 2013;49:13744.
38. Li Y, Pang H, Guo Z, Lin L, Dong Y, Li G, et
al. Interactions between drugs and polymers
influencing hot melt extrusion. J Pharm
Pharmacol. 2014;66(2):14866.
39. Kim NA, Choi DH, Lim JY, Kim KH, Lim DG,
Lee E, et al. Investigation of polymeric
excipients for dutasteride solid dispersion and
its physicochemical characterization. Arch
Pharm Res. 2014;37(2):21424.
40. Punčochová K, Ewing A V., Gajdošová M,
Pekárek T, Beránek J, Kazarian SG, et al. The
Combined Use of Imaging Approaches to
Assess Drug Release from Multicomponent
Solid Dispersions. Pharm Res. 2017;34(5):990
1001.
41. McFall H, Sarabu S, Shankar V, Bandari S,
Murthy SN, Kolter K, et al. Formulation of
aripiprazole-loaded pH-modulated solid
dispersions via hot-melt extrusion technology:
In vitro and in vivo studies. Int J Pharm.
2019;554(June 2018):30211.
42. Dong W, Su X, Xu M, Hu M, Sun Y, Zhang P.
Preparation, characterization, and in vitro/vivo
evaluation of polymer-assisting formulation of
atorvastatin calcium based on solid dispersion
technique. Asian J Pharm Sci. 2018;13(6):546
54.
43. Yu M, Ocando JE, Trombetta L, Chatterjee P.
Molecular Interaction Studies of Amorphous
Solid Dispersions of the Antimelanoma Agent
Betulinic Acid. AAPS PharmSciTech.
2015;16(2):38497.
44. Tres F, Coombes SR, Phillips AR, Hughes LP,
Wren SAC, Aylott JW, et al. Investigating the
dissolution performance of amorphous solid
dispersions using magnetic resonance imaging
and proton NMR. Molecules.
2015;20(9):1640418.
45. Alshahrani SM, Lu W, Park J-B, Morott JT,
Alsulays BB, Majumdar S, et al. Stability-
enhanced Hot-melt Extruded Amorphous Solid
Dispersions via Combinations of Soluplus® and
HPMCAS-HF. AAPS PharmSciTech.
2015;16(4):82434.
46. Smeets A, Koekoekx R, Clasen C, Van den
Mooter G. Amorphous solid dispersions of
darunavir: Comparison between spray drying
and electrospraying. Eur J Pharm Biopharm.
2018;130:96107.
47. Albadarin AB, Potter CB, Davis MT, Iqbal J,
Korde S, Pagire S, et al. Development of
stability-enhanced ternary solid dispersions via
combinations of HPMCP and Soluplus
®
processed by hot melt extrusion. Int J Pharm.
2017;532(1):60311.
48. Hu XY, Lou H, Hageman MJ. Preparation of
lapatinib ditosylate solid dispersions using
solvent rotary evaporation and hot melt
extrusion for solubility and dissolution
enhancement. Int J Pharm. 2018;552(12):154
63.
49. Li N, Taylor LS. Tailoring supersaturation from
amorphous solid dispersions. J Control Release.
2018;279:11425.
50. Volkova T V., Perlovich GL, Terekhova I V.
Enhancement of dissolution behavior of
antiarthritic drug leflunomide using solid
dispersion methods. Thermochim Acta.
2017;656:1238.
51. Zhang Q, Zhao Y, Zhao Y, Ding Z, Fan Z,
Zhang H, et al. Effect of HPMCAS on
recrystallization inhibition of nimodipine solid
dispersions prepared by hot-melt extrusion and
dissolution enhancement of nimodipine tablets.
Colloids Surfaces B Biointerfaces.
2018;172(May):11826.
52. Indulkar AS, Lou X, Zhang GGZ, Taylor LS.
Insights into the Dissolution Mechanism of
Ritonavir-Copovidone Amorphous Solid
Dispersions: Importance of Congruent Release
for Enhanced Performance. Vol. 16, Molecular
Pharmaceutics. 2019. 13271339 p.
53. Wlodarski K, Sawicki W, Kozyra A, Tajber L.
Physical stability of solid dispersions with
respect to thermodynamic solubility of tadalafil
in PVP-VA. Eur J Pharm Biopharm.
2015;96(August):23746.
54. Parikh T, Gupta SS, Meena AK, Vitez I,
Mahajan N, Serajuddin ATM. Application of
Film-Casting Technique to Investigate Drug-
M. E. T Butarbutar et al / Indo J Pharm 1 (2020) 55-68
68
Polymer Miscibility in Solid Dispersion and
Hot-Melt Extrudate. J Pharm Sci.
2015;104(7):214252.
55. Brandl M, Frank K, Westedt, Rosenblatt, Hölig,
Rosenberg, et al. The amorphous solid
dispersion of the poorly soluble ABT-102 forms
nano/microparticulate structures in aqueous
medium: impact on solubility. Int J
Nanomedicine. 2012;5757.
56. Ma X, Williams RO. Characterization of
amorphous solid dispersions: An update. J Drug
Deliv Sci Technol. 2019;50:11324.
57. Das SK, Kahali N, Bose A, Khanam J.
Physicochemical characterization and in vitro
dissolution performance of ibuprofen-Captisol®
(sulfobutylether sodium salt of β-CD) inclusion
complexes. J Mol Liq. 2018;261(2017):23949.
58. Das SK, Kahali N, Bose A, Khanam J.
Physicochemical characterization and in vitro
dissolution performance of ibuprofen-Captisol®
(sulfobutylether sodium salt of β-CD) inclusion
complexes. J Mol Liq. 2018;261(2017):23949.
59. Ha ES, Choo GH, Baek IH, Kim MS.
Formulation, characterization, and in vivo
evaluation of celecoxib-PVP solid dispersion
nanoparticles using supercritical antisolvent
process. Molecules. 2014;19(12):2032539.
60. Thakral NK, Thakral S, Stephenson GA,
Sedlock R, Suryanarayanan R. Compression-
Induced Polymorphic Transformation in
Tablets: Role of Shear Stress and Development
of Mitigation Strategies. J Pharm Sci.
2019;108(1):47684.
61. Homayouni A, Sadeghi F, Nokhodchi A,
Varshosaz J, Afrasiabi Garekani H. Preparation
and characterization of celecoxib solid
dispersions; comparison of poloxamer-188 and
PVP-K30 as carriers. Iran J Basic Med Sci.
2014;17(5):32231.
62. Zhu Q, Toth SJ, Simpson GJ, Hsu HY, Taylor
LS, Harris MT. Crystallization and dissolution
behavior of naproxen/polyethylene glycol solid
dispersions. J Phys Chem B. 2013;117(5):1494
500.
63. Homayun B, Lin X, Choi H-J. Challenges and
Recent Progress in Oral Drug Delivery Systems
for Biopharmaceuticals. Pharmaceutics.
2019;11(3):129.
64. Baird JA, Taylor LS. Evaluation of amorphous
solid dispersion properties using thermal
analysis techniques. Adv Drug Deliv Rev.
2012;64(5):396421.
65. Jójárt-Laczkovich O. Amorphization of a
crystalline active agent with the aim of
pharmaceutical technological formulation. 2012.
66. Wlodarski K, Sawicki W, Haber K, Knapik J,
Wojnarowska Z, Paluch M, et al.
Physicochemical properties of tadalafil solid
dispersions - Impact of polymer on the apparent
solubility and dissolution rate of tadalafil. Eur J
Pharm Biopharm. 2015;94(May):10615.
67. Wlodarski K, Sawicki W, Kozyra A, Tajber L.
Physical stability of solid dispersions with
respect to thermodynamic solubility of tadalafil
in PVP-VA. Eur J Pharm Biopharm.
2015;96(August):23746.
68. Rams-Baron M, Jachowicz R, Boldyreva E,
Zhou D, Jamroz W, Paluch M, et al. Physical
Instability: A Key Problem of Amorphous
Drugs. Amorphous Drugs. 2018. 107157 p.
69. Chavan RB, Rathi S, Jyothi VGSS, Shastri NR.
Cellulose based polymers in development of
amorphous solid dispersions. Asian J Pharm Sci.
2018;248264.