Anda di halaman 1dari 13

Review

Refining stability and dissolution


rate of amorphous drug
formulations
1. Introduction
Holger Grohganz, Petra A Priemel, Korbinian Lobmann,
2. Polymer-based solid
Line Hagner Nielsen, Riikka Laitinen, Anette Mullertz,
dispersions
Guy Van den Mooter & Thomas Rades
3. Physical separation
University of Copenhagen, Department of Pharmacy, Copenhagen, Denmark
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

4. Co-amorphous formulations
5. Recent trends
Introduction: Poor aqueous solubility of active pharmaceutical ingredients
(APIs) is one of the main challenges in the development of new small
6. Conclusion
molecular drugs. Additionally, the proportion of poorly soluble drugs among
7. Expert opinion new chemical entities is increasing. The transfer of a crystalline drug to its
amorphous counterpart is often seen as a potential solution to increase the
solubility. However, amorphous systems are physically unstable. Therefore,
pharmaceutical formulations scientists need to find ways to stabilise
amorphous forms.
Areas covered: The use of polymer-based solid dispersions is the most
established technique for the stabilisation of amorphous forms, and this
review will initially focus on new developments in this field. Additionally,
newly discovered formulation approaches will be investigated, including
For personal use only.

approaches based on the physical restriction of crystallisation and crystal


growth and on the interaction of APIs with small molecular compounds rather
than polymers. Finally, in situ formation of an amorphous form might be an
option to avoid storage problems altogether.
Expert opinion: The diversity of poorly soluble APIs formulated in an
amorphous drug delivery system will require different approaches for their
stabilisation. Thus, increased focus on emerging techniques can be expected
and a rational approach to decide the correct formulation is needed.

Keywords: amorphous, co-amorphous systems, dissolution enhancement, in situ,


microcontainers, polymers, solid dispersions, stability, surface crystallisation

Expert Opin. Drug Deliv. (2014) 11(6):977-989

1. Introduction

An increasing number of active pharmaceutical ingredients (API) suffer from one


common challenge: their low solubility in water. These drugs are classified as
class 2 and class 4 in the biopharmaceutics classification system (BCS), respectively [1].
While in the period of 2000 -- 2011, only around 20% of generics approved via the
abbreviated new drug applications system in the USA belonged to BCS class 2 [2], up
to 90% of the APIs currently under development are classified as BCS class 2 or
class 4 drugs [3]. Low aqueous solubility can seriously reduce the bioavailability of
the drug [4], and the further development of pharmacologically effective new chemi-
cal entities might be abandoned if the solubility issue cannot be resolved satisfactory.
The transformation of an API from its crystalline state to an amorphous form is gen-
erally regarded as an option to handle this challenge. Amorphous materials lack the
three dimensional long-range positional and orientational order of molecules typical
for crystalline solids and represent the highest energetic state of a solid material [5].
Amorphous drugs are expected to be advantageous in terms of solubility and dissolu-
tion rate because one of the determining steps in the dissolution process of crystalline

10.1517/17425247.2014.911728 2014 Informa UK, Ltd. ISSN 1742-5247, e-ISSN 1744-7593 977
All rights reserved: reproduction in whole or in part not permitted
H. Grohganz et al.

dispersions has been provided by Chiou and Riegelman [9],


Article highlights. but currently, the term solid dispersion is mostly linked to
(supersaturated) glass solutions of poorly soluble compounds
. Poor aqueous solubility is a key challenge for
pharmaceutical development. with amorphous or semicrystalline polymeric carriers.
. Amorphous systems offer an increase in dissolution rate A glass solution in the true sense is a one-phase system in
and solubility but at the cost of inherent instability. which all molecules of the API are intimately mixed with
. Polymer-based solid dispersions are well-described the carrier molecules. The selection of the carrier is important
systems for stabilisation of an amorphous formulation. as it will have significant impact on the physical stability and
. Physical separation may restrict crystallisation or
crystal growth. dissolution performance of the solid dispersions. An overview
. A promising new approach for stabilisation is of the polymers that are being used in marketed formulations
co-amorphous systems, which are based on is provided in a recent review [10]. Polyethylene glycols,
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

small-molecular-weight components. hydroxypropyl methylcellulose (HPMC) and polyvinylpyrro-


. The issue of low storage stability can potentially be lidone (PVP) have been most often used in the past, whereas
avoided by in situ amorphisation.
more recently, HPMC acetate succinate has been frequently
This box summarises key points contained in the article. applied because it shows to be successful in maintaining
supersaturation of dissolved API [11,12]. Also, the amphiphilic
graft copolymer of polyethylene glycol, polyvinylcaprolactam
solids, the disruption of the crystal lattice [6], does not have and polyvinylacetate (Soluplus) is a promising carrier due to
to take place. However, the drawback of the increased free its good flowability and excellent extrudability [13].
energy of amorphous material is its inherent thermodynamic The ease of crystallisation of a drug from its amorphous state
instability leading to relaxation, nucleation and crystal growth depends on the driving force for crystallisation given by the free
phenomena during storage [7]. Also, precipitation and crystal- energy difference between the amorphous and the crystalline
lisation during dissolution in the gastrointestinal fluids can state and between the mobility of the amorphous state and the
occur. Pure amorphous drugs are, therefore, usually not devel- crystallisation mechanism. In order to decrease pill burden,
For personal use only.

oped as such to commercial dosage forms but in combination one aims to have API loadings as high as possible in the carrier.
with excipients that stabilise the amorphous form during In many cases, API loadings up to 30 -- 40% can be obtained
storage and prevent nucleation and crystallisation during the without noticeable phase separation (amorphous--amorphous
dissolution process. phase separation or the presence of a crystalline API phase)
The stabilisation of amorphous APIs via various formula- immediately after preparation. However, phase separation as a
tion approaches is therefore a highly investigated area in function of time and/or triggered by storage conditions
pharmaceutical research. Due to the increasing numbers and (temperature and/or elevated humidity) is often reported and
thus increasing diversity of poorly water-soluble APIs, there contributes to the large discrepancy between the scientific and
is a need for an increasing amount of different technological developmental efforts and the low number of solid dispersions
approaches to stabilise amorphous formulations [8]. Solid that have entered the market. The solubility of crystalline drugs
dispersions are the most thoroughly investigated approach so in polymeric matrices is of importance for selecting the appro-
far, and in this review, we will start with a focus on recent priate polymer because it will establish the limit of drug loading.
developments in polymer-based solid dispersions (Section 2). The high polymer viscosity hampers straightforward solubility
However, polymer-based systems may not be applicable determination and is one of the reasons that there is still
in all cases and generally require a rather large amount of no universally accepted technique to measure the solid state
excipient. This has led to a recent increase in research aiming drug--polymer solubility/miscibility. Vasanthavada et al. there-
to find new ways of stabilising amorphous drug formulations. fore proposed to estimate the solubility of trehalose in solid
Physical separation principles have shown to be beneficial in dispersions with dextran or PVP by measuring the water-
reducing crystallisation or restricting crystal growth and are vapour-induced crystallisation of trehalose [14]. Recently,
discussed in Section 3. Apart from polymers, also small Mahieu et al. developed a new protocol to determine the drug
molecules, both API and excipients, recently showed a poten- solubility in polymeric matrices, based on the idea that the
tial to increase the stability of amorphous formulations equilibrium-saturated state is reached faster by demixing of
(Section 4). Finally, recent trends such as stable spray-dried supersaturated solid solutions compared to dissolution of
salts or in situ formation of an amorphous system may prove crystalline drugs, as measured by melting point depression
valuable formulation options for poorly water-soluble drugs studies [15,16]. The equilibrium solubility was thus determined
(Section 5). by following the precipitation of indometacin (IMC) from a
PVP matrix when annealed above the glass transition tempera-
2. Polymer-based solid dispersions ture (Tg). A theoretical frame regarding the thermodynamic
basis of phase separation as a result of supersaturation in solid
The combination of an API with excipients is known as a dispersions has been proposed [17]. Solid dispersions of felodi-
solid dispersion. A classification of the different types of solid pine and nifedipine with PVP were miscible over the whole

978 Expert Opin. Drug Deliv. (2014) 11(6)


Refining stability and dissolution rate of amorphous drug formulations

composition range. However, based on the Flory-Huggins common setup, a powder blend is introduced via a hopper
(FH) lattice theory, the predicted solid solubility value for into a heated barrel with a rotating screw, where the powder
nifedipine was < 10% w/w. Based on these findings, the authors blend is intensively mixed in the liquid state and moved
hypothesised that for most miscible drug -- polymer glass solu- towards a die that shapes the melt as strands, films, pellets or
tions, physical stabilisation will play a large role. Similar differ- tablets. The amount of heat and shear forces applied, as well
ences between calculated solid state solubility and practical as the rate of cooling when the extrudate leaves the die,
values were reported for naproxen-PVP and itraconazole- contributes to the physical structure of the solid dispersion.
eudragit E100 systems [18,19]. The increasing interest in the A relatively new pharmaceutical processing technology is the
FH-based model witnessed through recent literature is encour- KinetiSol Dispersing technology, which is a high-energy mix-
aging, although the focus remains on the modification of the ing process for the production of amorphous pharmaceutical
classical analytical expressions [20-22]. Quantitative thermody- solids [30]. This technology utilises a series of rapidly rotating
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

namic values of the strength of specific interactions such as blades to process the drug and polymeric carrier through a
hydrogen bonding and the degree to which they influence ther- combination of kinetic and thermal energy without external
modynamic properties still remain unaccounted in these misci- heating sources. The mild conditions at which carbon dioxide
bility models for interacting systems. The use of other modified can exist as a supercritical fluid (31.06 C at 73.8 bar)
forms of the FH model that account for specific interactions prompted researchers to exploit this material as processing
between mixing components might be advisable. One ubiqui- aid during manufacturing. Solid dispersions of furosemide
tously used methodology in polymer chemistry for the study and carbamazepine (CBZ) were prepared using supercritical
of miscibility behaviour of hydrogen bonding blends is the carbon dioxide as antisolvent [31,32]. Verreck et al. studied the
Painter and Coleman association model [23]. For the expression possibility to apply supercritical and subcritical carbon dioxide
of DGmix, this model includes a specific term of Gibbs free as a temporary plasticiser during hot-melt extrusion [33,34].
energy change due to hydrogen bonding in addition to the
combinatorial entropy and enthalpy of mixing. Its magnitude 3. Physical separation
depends on the relative strength of self-associating to inter-
For personal use only.

associating hydrogen bonding and the density of specific Another strategy to improve the physical stability of the amor-
interacting sites. Another useful, yet unexploited, model in phous form of a drug is to separate the amorphous drug in
pharmaceutical sciences is the Wertheim thermodynamic microcontainers and thereby restrict the crystal growth of
perturbation theory, which accounts for directional hydrogen the drug to the diameter of the microcontainer. Microcon-
bonding interactions [24]. tainers are small polymeric devices consisting of a flat base
As glass solutions are often supersaturated, kinetic stabilisa- surrounded by a walled reservoir and were fabricated in three
tion plays an important role and the manufacturing process different sizes of 73, 174 and 223 m in inner diameter [35,36].
will significantly contribute to the degree of supersaturation. Amorphous IMC was prepared in the three different sizes of
The two most applied manufacturing methods for solid disper- microcontainers by melting g-IMC in an oven, followed by
sions in pharmaceutical industry are spray drying and hot-melt quench-cooling. The samples were stored at 30 C and 23%
extrusion. The former process starts from a solution of API and RH for up to 30 days. Raman microscopy was used to observe
carrier in a common organic solvent or mixture of solvents, if each microcontainer contained amorphous or crystalline
which is subsequently atomised using a nozzle and quickly IMC over time, and numeric crystallisation was obtained
evaporated to obtain a powder. The very fast solvent evapora- and compared with the crystallisation rate of amorphous
tion contributes to the amorphous state of the formed solid bulk IMC [35].
dispersion. Janssens et al. reported the importance of fast The onset of crystallisation for the amorphous bulk IMC to
solvent evaporation for miscibility. Solid dispersions made up the crystalline form occurred rapidly, and around 51%
of itraconazole and Eudragit E100 were better mixed follow- converted within 30 days of storage (Figure 1). Improvement
ing spray drying compared to a film-casting process with a low in stability was seen when confining amorphous IMC in the
rate of solvent evaporation [18]. Recent findings also indicate microcontainers, but different levels of crystallinity between
the importance of solvent characteristics in determining the the two larger sized microcontainers were observed. After
properties of the resulting solid dispersions [25,26]. Alternatively, day 30, the numeric crystallisation was found to be 29 and
solid dispersions can be prepared by spraying a drug--polymer 38% in the 174 m and 223 m microcontainers, respec-
solution onto inert beads. A layer of solid dispersion is thus tively. In the 73 m microcontainers, no stability improve-
formed following evaporation of the organic solvent [27]. The ment was observed as no remaining amorphous IMC was
commercial form of itraconazole (Sporanox) is an example found after 15 days of storage. This unexpected behaviour
of this process. Spray congealing where a melt is transformed may be due to the material used to fabricate the microcon-
into solid spherical particles by spraying the melt into a cooling tainers (SU-8), as it is known to absorb moisture or capillary
chamber is a solvent-free variant of spray drying [28]. Hot-melt condensation in the small microcontainers [35].
extrusion is a scalable and industrially applicable technology The confined amorphous IMC contains cracks in the
originating from the polymer industry [29]. In the most surface, and these cracks were proposed to be nucleation sites

Expert Opin. Drug Deliv. (2014) 11(6) 979


H. Grohganz et al.

60 Amorphous bulk indomethacin

Crystalline indomethacin (%)


Oven/N2
Heating plate/N2 (cooling rate: 36 K/min)
40 Heating plate/ice (cooling rate: 23 K/min)
Heating plate/room temperature (cooling rate: 14 K/min)

20

0
Preparation conditions
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

Figure 1. Numeric crystallisation for amorphous bulk IMC and amorphous IMC confined in 223 mm microcontainers prepared
either by melting the IMC in an oven and cooling with liquid nitrogen or melting on a heating plate and cooling either at a
rate of 36, 23 or 14 K/min. The samples were stored at 22 C and 33% RH for 30 days and are an average of three replicates.

for crystal growth. Therefore, it was investigated if the prepa- Acryl-EZE started to recrystallise at the surface shortly after
ration conditions of the amorphous IMC could have an effect preparation [41]. Nevertheless, as the crystalline and the amor-
on the physical stability. The largest microcontainers were phous form of the drug are highly soluble, the dissolution
filled with g-IMC, melted on a heating plate and cooled behaviour of the extrudates was not affected by the solid state
with either liquid nitrogen (36 K/min), ice (23 K/min) or at change.
room temperature (14 K/min). Quench-cooling led to a Due to the importance of surface crystallisation, approaches
significant risk of surface cracking, while cooling the melt of were undertaken to prevent or inhibit surface crystallisation.
For personal use only.

IMC at 14 K/min resulted in a smooth surface without any Amorphous IMC, griseofulvin and nifedipine were coated
cracks. The physical stability of the microcontainer-confined with a thin gold layer, and all drugs exhibited reduced surface
amorphous IMC was again investigated using Raman micros- crystallisation [42-44]. A pharmaceutically more relevant formu-
copy. The fastest cooled IMC (36 K/min) had a crystallisation lation was investigated using physical mixtures of amorphous
number of about 31% after 30 days of storage, whereas the IMC and either Eudragit E or Soluplus particles [45]. IMC
melt of IMC cooled at 23 K/min and 14 K/min had numeric crystallisation was slowed down by the addition of Eudragit
crystallisation of around 21 and 10% after 30 days of storage, E but not by Soluplus particles. Scanning electron microscopy
respectively (Figure 1), indicating that the more gentle cooling revealed surface coverage of IMC by Eudragit E particles
conditions reduced the likelihood of crystallisation [36]. It was (Figure 2). In contrast, a simple mixture formed when combin-
concluded that microcontainers can be utilised for stabilising ing amorphous IMC particles with Soluplus particles. Reduced
amorphous IMC and that the preparation conditions affect molecular mobility through surface coverage and mechanical
the physical stability of amorphous IMC. hindrance of outwards growing crystallites are thought to
Besides restricting the surface by confinement in physical slow down surface crystallisation [46]. The surface crystallisation
containers, also the surface of a particle in itself has to be of the above-mentioned hot-melt extrudates of guaifenesin
considered. The particle surface separates the particle from could be slowed down after they were coated with Aquacoat
its environment and has considerably different properties ECD but not when Opadry was used as coating agent [47].
compared with the bulk material. Surface properties have The latter polymer has a lower solubilisation potential for the
often been neglected as their contribution to the overall prop- drug and additionally requires a higher curing temperature,
erties of a material is relatively small [37]. However, molecules hence molecular mobility of drug molecules increased and
situated at the surface have a higher molecular mobility than led to crystallisation.
the bulk molecules [38,39] and surface crystallisation of amor- Recently, different stabilisation approaches for amorphous
phous material is thought to be a consequence thereof. Ana- spin-coated films were compared to each other, namely a glass
lytically, surface crystallisation may result in different solution film, a drug film coated with a polymer film and a
quantitative crystallinity values, depending on the analytical drug film on top of a polymer film [48]. The study was
technique used (i.e., if the analytical method has a surface performed using CBZ, felodipine, fenofibrate and celecoxib
bias or not). Most importantly, surface crystallisation was as model drugs and PVP K30, Eudragit E PO and Soluplus
found to reduce the dissolution behaviour of amorphous as polymers. In glass solutions, drug molecules are immobi-
IMC in such a way that the amount of dissolved drug was lised by the polymer chains, the Tg is raised and molecular
the same as the crystalline form [40]. However, a change in interactions between drug and polymer may be established.
dissolution behaviour is not always observed. Hot-melt Surface coverage reduces surface molecular mobility, and the
extrudates of guaifenesin with either Eudragit L100-55 or drug film or particle is separated from its environment. Under

980 Expert Opin. Drug Deliv. (2014) 11(6)


Refining stability and dissolution rate of amorphous drug formulations

for more than 6 months when stored at 40 C/75% RH,


whereas amorphous IMC alone recrystallised after 1 month [57].
In addition, the pSi carrier facilitated immediate release of IMC
and enhanced oral delivery performance in in vivo absorption
study. A 200% Cmax was reached four times faster (i.e., shorter
Tmax), and a significant increase in bioavailability were observed
compared to crystalline IMC and a commercially available
formulation.

4. Co-amorphous formulations
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

The increasing need to improve the dissolution properties of


poorly water-soluble drugs and the interest in combining drugs
for more efficient medication have led to the development of
co-amorphous drug--drug formulations. This novel formula-
tion approach offers the opportunity not only to enhance
Figure 2. Scanning electron microscopy picture of indome-
bioavailability of BCS class 2 drugs but also to prepare formu-
tacin with surface coverage by Eudragit E. lations intended for combination therapy. Moreover, reduc-
tion in the size of the dosage form compared to the
traditional drug--polymer glass solutions, where generally large
high humidity conditions, the polymer-coated drug films polymer amounts are used, can be regarded as an additional
were physically more stable than the glass solution films and advantage.
the drug film on polymer-coated substrate. Hence the reduced The concept of co-amorphous formulations was intro-
surface crystallisation and the protection from humidity were duced by Chieng et al. [59] to differentiate amorphous mixtures
For personal use only.

more effective in stabilising the amorphous nature of the drug of two low-molecular-weight components from amorphous
at high humidity. However, this order might change if the solid dispersions of a drug and a polymer. In the initial study,
study was performed at lower humidity, where plasticisation the two drugs IMC and ranitidine HCl were combined with
of the glass solution and potential phase separation are less the aim to enhance their complimentary pharmacological
prominent, as previously reported for some glass solutions [49]. effect. More specifically, by administering the histamine
The use of mesoporous inorganic materials (porous silicon, H2-receptor antagonist ranitidine HCl in an amorphous com-
silica, Al2O3, TiO2, carbon and hydroxycarbonate apatite) is bination with NSAID IMC, the gastrointestinal side effects
another relatively new approach to overcome poor drug solu- associated with NSAIDs may be reduced. Since then, a similar
bility via amorphisation and physical separation [50-52]. Incor- combination of a poorly soluble NSAID and an antihistaminic
poration of a drug into mesoporous materials for example, by drug (naproxen-cimetidine) [60] in addition to combinations of
solvent deposition [52,53], mechanical activation (co-grinding two NSAIDs (naproxen-IMC) [61] and two drugs intended for
or milling) [54] or vapour-phase-mediated mass transfer [55] the treatment of metabolic disorders (simvastatin-glipizide) [62]
may lead to loss of the crystalline structure of the adsorbed have been investigated.
drug molecules. Nucleation and crystal growth of the drug in The drug--drug combinations, generally processed by
the pores are hindered by spatial constraints as long as the melt-quenching or mechanical activation in molar ratios of
pores are smaller than the critical nucleus of the drug, which 2:1, 1:1 and 1:2, formed single-phase amorphous systems, as
makes the nucleus growth energetically unfavourable [50-52]. indicated by single, composition-dependent Tg. However,
In small pores (< 10 nm wide), drugs have been observed to in co-amorphous drug--drug systems the formulation with
be in the amorphous state [53,56]. In case the pore and the the highest Tg did not usually turn out to be the most stable
drug cluster in the pore are larger than the critical nucleus, mixture. Thus, an increase in Tg alone cannot explain the
crystallisation may occur but it can still be prevented by kinetic principal stabilisation mechanism. Applying spectroscopic
factors if the rate of nucleation is low [50]. However, if the drug methods, specific hydrogen-bonding or p--p interactions
clusters within the material are physically separated from each between the two molecules were shown to lead to superior
other (i.e., in non-interconnected pores), nucleation takes place stability of the 1:1 combinations. For example, the 1:1
independently within each pore. Thus, in order to a significant co-amorphous mixtures of IMC-ranitidine HCl, naproxen-
amount of loaded drug to crystallise, a high nucleation rate is cimetidine and naproxen-IMC remained physically stable,
required. Due to this stabilising mechanism, several drug- whereas the two other blends (1:2 and 2:1) showed recrystal-
loaded mesoporous systems have been observed to be lisation of the excess component in the stability studies [59-61].
physically stable under accelerated storage conditions [54,57,58]. The intermolecular interactions were also shown to be an
For example, amorphous IMC-pSi-ox (thermally oxidised important dissolution-enhancing factor in co-amorphous
porous silicon) systems (pSi-ox) were observed to be stable mixtures. Not only was the dissolution rate increased above

Expert Opin. Drug Deliv. (2014) 11(6) 981


H. Grohganz et al.

the individual crystalline and amorphous drugs, but it was the initially requested demands. First, the prepared blends
also synchronised for the two components [60,61]. In the case were homogeneous, molecular-level mixtures. Second, specific
of naproxen-IMC, intermolecular hydrogen bonding was molecular interactions between the drug and the amino acids
observed to result in the formation of an energetically stable could be identified, both hydrogen bonds and p--p interactions.
heterodimer between naproxen and IMC [63]. In order for Third, all co-amorphous blends had an increased Tg compared
recrystallisation to occur, the hydrogen bonds within the het- to the pure drugs Tg. In addition, several drug--amino acid
erodimer first need to be broken. Subsequently, the molecules combinations had Tg values in the range of 70 -- 80 C, which
need to rearrange themselves and establish interactions is in the generally desired range for Tg in amorphous formula-
between like molecules in order to recrystallise as homo- tions, assuming storage conditions at room temperature (Tg
dimers. This cascade is assumed to reduce the likelihood of minus 50 K rule) [66]. Because of these findings, it was not
re-crystallisation. surprising that seven out of the eight co-amorphous mixtures
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

The formation of intermolecular hydrogen bonds is, how- remained physically stable over at least 1 year at room tempera-
ever, not a strict prerequisite for an increase in stability. It ture and at 40 C, under dry storage conditions (Figure 3) [67]. By
has been shown that molecular-level mixing may be sufficient contrast, the pure amorphous drugs, CBZ and IMC, showed
to obtain an enhanced stability of co-amorphous mixtures [62]. recrystallisation within 1 week at both storage conditions. The
Single-phase mixtures of simvastatin and glipizide showed only co-amorphous blend that did not remain amorphous
increased physical stability compared with physical mixtures over a period of 1 year at both storage conditions was IMC-
of the corresponding amorphous drugs, even though no inter- PHE. This blend showed recrystallisation of both IMC and
molecular interactions were observed in the co-amorphous PHE after a period of 6 months at 40 C. This mixture had
mixtures. However, the lack of specific interactions was the lowest Tg of all investigated blends and in addition, did
reflected in the fact that the dissolution rate did not improve not show any signs of molecular interactions. This supports
compared with the individual amorphous drugs. the idea of a stabilising interplay of molecular level mixing,
It can be concluded that the strength of the co-amorphous molecular interactions and increased Tg. Finally, the co-
drug--drug systems lies in their ability to offer stabilisation of amorphous drug amino-acid mixtures were tested towards their
For personal use only.

the amorphous forms at a lower bulk volume compared to intrinsic dissolution rate (IDR) in phosphate buffer (pH 7.2).
amorphous polymer dispersions and a potential for synchron- All formulations had an increased IDR compared with the
ised release of the two APIs. However, this approach should pure crystalline and amorphous drugs.
not be limited to combinations of two APIs, as this would In addition to amino acids as low-molecular-weight stabil-
be limited by pharmacologically meaningful combinations. isers for the amorphous drug, there is a growing interest of
Accordingly, inactive low-molecular-weight excipients were using other low-molecular excipients to stabilise drugs in the
combined with a poorly soluble drug, thus expanding the amorphous form. These include weak carboxylic acids, such
co-amorphous concept. From the studies above, it became as citric acid [68] and tartaric acid [69], weak bases, such as
apparent that the investigated excipients should ideally result meglumine [70], saccharin [71], phospholipids [72], sugars,
in directed molecular interactions, molecular level mixing urea and nicotinamide [73]. The stabilisation mechanism of
and if possible, an increase of the drug Tg. Considering the these co-amorphous formulations was again attributed mainly
interaction of APIs with the active receptor site in the human to molecular or ionic interactions (salt formation) between the
body, amino acids were chosen as a rational starting point. drug and excipient in the blend, but also an increase in drug
In two studies by Lobmann et al. [64,65], the amino acids Tg in some cases. In a particular co-amorphous mixture, a
arginine, phenylalanine (PHE), tryptophan and tyrosine co-precipitated blend of acyclovir and citric acid at a molar
were investigated towards their suitability as co-amorphous ratio of 1:2 offered in addition to the amorphous stabilisation
formation agent, stabiliser and dissolution enhancer in combi- also an increased improvement of skin permeation flux com-
nation with APIs CBZ and IMC. The authors were able to pared with crystalline acyclovir [74]. In another study, even
prepare several different co-amorphous mixtures with one or at storage conditions above the Tg, the co-amorphous blend
two amino acids at the molar ratios 1:1 or 1:1:1. From the containing paracetamol and citric acid showed high physical
preparative ball milling studies, 8 of 16 investigated mixtures stability for at least 2 years due to the strong intermolecular
formed a single, co-amorphous phase. It became apparent that interactions [75].
the four investigated amino acids were not equally suitable to Overall, co-amorphous formulations proved to be a
induce the formation of co-amorphous mixtures with the promising new approach and alternative to polymer solid dis-
drugs. In this context, tryptophan showed very good amorph- persion and the future will show if this new and promising
isation properties, arginine and PHE showed intermediate approach can live up to its expectations.
amorphisation properties for co-amorphous systems, whereas
tyrosine was unable to form any co-amorphous phase in any 5. Recent trends
of the investigated mixtures with the drugs.
During further investigations, most of the successfully In the previous section, it was shown that molecular interac-
prepared co-amorphous drug amino-acids mixtures fulfilled tions by for example, hydrogen bonds or p--p interactions

982 Expert Opin. Drug Deliv. (2014) 11(6)


Refining stability and dissolution rate of amorphous drug formulations

non-salt amorphous form [80]. Poorly water-soluble sulfathia-


zole turned out to be amorphous after spray drying from an
ethanol/water solution, whereas pure ethanol resulted in crys-
talline sulfathiazole, showing the influence of the solvent [79].
Furthermore, sulfathiazole sodium salts were produced using
various solvents (both aqueous and organic) and no crystalli-
sation was observed over a period of 12 months when stored
CBZqc
at dry conditions at 4 C. The salts were not stable at ambient
conditions (18 -- 22 C and 40 -- 80% RH), and the non-salt
CBZ-TRP
forms of amorphous sulfathiazole started to crystallise already
CBZ-PHE-TRP after 1 h regardless of the storage conditions [79]. The stability
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

was improved significantly by co-spray drying solutions of


Intensity

CBZ-ARG-TRP
sulfathiazole and sulfathiazole sodium. The optimal ratio
IMC-PHE-TRP between the acid and salt was found to be 1:1, which led to
IMC-ARG-PHE at least 6 months stability at ambient conditions, but even
small amounts of salt increased the stability significantly [81].
IMC-ARG
The varying oral bioavailability of the BCS class 4 drug furo-
IMC-TRP semide is closely related to its dissolution rate [82,83]. It was
5.7 PHE
possible to produce amorphous furosemide sodium salt and
IMC-PHE amorphous furosemide acid by spray drying [84]. The amor-
phous salt had a Tg 39 C higher than the amorphous
acid [84]. The amorphous salt showed promising in vitro
characteristics in terms of higher solubility and dissolution
rate in physiologically relevant media and a significantly faster
IMC
For personal use only.

Tmax (oral dosing to rats) compared with amorphous and


5 10 15 20 25 30 35 crystalline furosemide acid. Importantly, the amorphous salt
Angle/2q () was stable for 291 days at 22 C and 33% RH, whereas the
amorphous furosemide acid was only stable for 4 days at the
same conditions (Figure 4) [84]. At high humidity and during
Figure 3. XRPD diffractograms of co-amorphous drug
amino-acid mixtures and the respective pure drugs carba-
dissolution, the amorphous furosemide salt converted quickly
mazepine (CBZ) and IMC stored over a period of 1 year at to a trihydrate [85]. Spray drying furosemide together with
40 C/dry conditions. The diffractogram shown for co- HPMC stabilised the amorphous furosemide sodium salt
amorphous IMC-phenylalanine (PHE) was collected after a during dissolution at pH 6.5 [86].
storage period of 6 months and indicates recrystallisation of A very recent approach to address low storage stability of
both, IMC and PHE. The diffractograms of the pure amorphous formulations is the formation of an amorphous
amorphous drugs, IMC and CBZqc (quench-cooled), were form in situ. This was realised during intrinsic dissolution
collected after a storage time of 7 days and indicate the fast testing for crystalline g-IMC in the presence of Eudragit
recrystallisation of the individual drugs. E [87]. In situ amorphisation was observed when drug polymer
Adapted with permission from [67].
IMC: Indometacin.
compacts were immersed in a buffer of a pH value at which
the drug, but not the polymer, can dissolve, in this case at
contribute to an enhanced stability of an amorphous mixture. pH 6.8. Compacts changed colour from white to yellow,
If achievable, the formation of amorphous salts is another indicating amorphisation of IMC. DSC thermograms showed
option for molecular interaction. Spray drying is a well- a single Tg comparable to the glass solution of the same com-
known method for converting drugs to their amorphous position. In the scores plot of a principal component analysis
form in a reproducible manner [76,77] and is a suitable method of FT-ATR-IR spectra, in situ amorphised samples clustered
for producing amorphous salts, which often show a higher together with a quench-cooled glass solution of the same
stability than their respective non-salts [78,79]. The selection drug--polymer composition. In XRPD measurements, the
of solvent and spray-drying conditions (pressure, drying gas intensity of the reflections decreased considerably. Neverthe-
and temperature) influences the stability of the formed less, reflections did not disappear completely and scanning
amorphous drug [79]. electron microscopy was able to depict a few remaining
Amorphous sodium and potassium salts of the poorly crystallites in the cross-section of compacts after in situ
soluble drug chlorothiazide were prepared by spray drying amorphisation.
from both aqueous and organic solvent [77]. All chlorothiazide The mechanism of in situ amorphisation is assumed to be
salts were physically stable for at least 6 months when stored due to pH-dependent dissolution and ionisation behaviour.
at dry conditions at 25 C -- significantly longer than the The drug can partially dissolve, whereas the polymer is

Expert Opin. Drug Deliv. (2014) 11(6) 983


H. Grohganz et al.

Amorphous salt,
291 days storage

Amorphous salt,
4 days storage

Intensity (a.u.)

Amorphous acid,
4 days storage
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

Crystallline acid

10 20 30
Angle/2q ()

Figure 4. XRPD diffractogram of amorphous furosemide salt and acid after storage at 22 C and 33% RH. For comparison the
diffractogram for crystalline furosemide is also shown.

plasticised by the water and swells, enabling molecular mixing different materials were screened for their precipitation
to some extent. At a drug--polymer ratio of 3:1 and 1:1, the inhibition of nine drugs in a model fasted duodenal fluid [12].
For personal use only.

compacts swelled and were yellow throughout the whole Other groups used principal component analysis to gain
cross-section. However, the 1:3 ratio compact did not swell understanding about the effect of molecular properties of
to the same extent and amorphisation took place only on the polymers on drug precipitation behaviour [92] or screening
the surface, probably due to restricted penetration depth of combined with dissolution, interaction and imaging assays
buffer into the compact once the polymer has swollen. At with a strong focus on development of successful glass
pH 6.8, amorphisation did not improve dissolution behaviour solution formulations [93]. All these studies are important to
compared to a physical mixture. Dissolution advantages understand and optimise amorphous formulations with
became obvious at pH values at which the polymer dissolves. regard to their supersaturation and precipitation behaviour.
The 3:1 and 1:1 in situ samples out-performed the physical However, in a study where absorption was studied using
drug--polymer mixtures as well as a quench-cooled glass solu- CaCo cells, the effect of the precipitation inhibitor HPMC
tion. By contrast, at the 1:3 ratio, where only the surface was was found to be lower than estimated from studies in a non-
converted in situ, the glass solution was superior. absorptive environment [94]. Similarly, studies performed in
The dissolution behaviour of amorphous formulations has dogs [95] and humans [96] that precipitation in vivo might be
been extensively studied. The preparation technique used for overestimated from in vitro studies.
conversion into the amorphous form has been found to influ-
ence the dissolution rate of amorphous drugs such as IMC [88] 6. Conclusion
as well as glass solutions [89,90]. Due to their high-energy form
and increased dissolution rate, amorphous formulations will The transformation of drugs to an amorphous form is one of
create supersaturation upon dissolution. the key techniques to address the problem of low solubility of
However, supersaturation is thermodynamically unstable APIs. Several techniques are available to pharmaceutical scien-
and will usually lead to precipitation. Therefore, different tists, with polymer-based solid dispersions being the most
approaches to maintain supersaturation and inhibit precipita- investigated system. Here, the miscibility of the API with
tion have been investigated. By creating supersaturation in a the polymer is of importance and theoretical approaches can
slow manner, the maximum concentration of drug was lower assist in the choice of the right polymer. New developments
and was reached later. Nevertheless, the AUC in a concentra- are the application of physical separation by micro-containers
tion versus time profile was larger compared with fast creation and surface coverage, which have been shown to reduce
of supersaturation. This finding held true for drugs and for crystallisation or crystal growth. Exchanging polymers by
drug polymer glass solutions [91]. As already mentioned in small-molecular-weight substances led to the development of
Section 2, the polymer used for glass solution systems affects co-amorphous systems. Initially intended for combination
the dissolution rate and may inhibit precipitation. In order therapy with two drug molecules, the concept has now been
to find the most potent precipitation inhibitor for a drug, generalised by using amino acids as excipients. Finally, the

984 Expert Opin. Drug Deliv. (2014) 11(6)


Refining stability and dissolution rate of amorphous drug formulations

use of a crystalline system that undergoes in situ amorphisa- Another area of great practical importance in the develop-
tion close to application may circumvent the low stability of ment of amorphous formulations is the lack of reliable and
amorphous formulations. It can be summarised that prepara- reproducible stress tests with respect to their physical and
tion and stabilisation approaches for amorphous systems chemical stability. With the amorphous form being inherently
will play a major role in pharmaceutical sciences also in the non-Arrhenius in its behaviour, the prediction of physical
future, in order to address the growing challenge of poorly stability remains a challenge. In this context, the fast and
water-soluble APIs. reliable determination of the thermodynamic solubility of a
drug in a polymer remains challenging, despite recent
7. Expert opinion advancements in this field. Also, the influence of small
amounts of moisture on the solubility of the drug in a given
Over the last decades, formulation of poorly water-soluble polymer needs to be determined in a more predictable way.
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

drugs as amorphous systems, especially by using polymers as An intriguing development is the use of drug formulations
excipients, has made tremendous progress and several amor- that only convert to an amorphous form immediately before
phous drug formulations have successfully entered the market. administration or even in the gastrointestinal tract. This
This progress was based on a better understanding of the work is only at its very beginning, but recently it has been
nature of the amorphous form and the development of pre- found that precipitation of the drug during lipolysis of
parative techniques that are scalable in the pharmaceutical lipid-based drug delivery systems may occur in an amorphous
industrial context. However, still the number of amorphous form during biorelevant in vitro dissolution tests [97]. In this
drug formulations on the market is small, especially when form, precipitation of the drug may not constitute a formula-
considering the large number of poorly water-soluble drugs tion problem, but may still lead to a high bioavailability due
coming out of the drug discovery process and many challenges to the amorphous nature of the precipitate and thus a fast
still remain to be addressed before amorphous drug formula- re-dissolution that could be expected at the absorptive sites
tions can be routinely used in the development of medicines. in the small intestine. In fact, initial in vivo investigations
This review has shown that alternative or additional appear to support this conclusion [98,99].
For personal use only.

approaches to using drug polymer glass solutions merit fur- Several options were shown to be available for the solubility
ther investigation. Coating of amorphous forms, including improvement of poorly water-soluble drugs and for the stabi-
coating of glass solutions, may increase the stability of the lisation of amorphous systems. It can be expected that the
drug against crystallisation, by reducing surface mobility and trend of developing new approaches will continue and that
preventing or reducing water absorption into the amorphous several new techniques will emerge in the not too distant
formulation during storage. But just how to coat amorphous future. Considering the increasing diversity of poorly soluble
formulations in a reliable and scalable process requires further APIs, these different approaches will also be necessary for
research. Also, the segmentation of amorphous formulations optimal performance of the formulation. What still remains
into small, independent units can further stabilise the formu- to be achieved is the establishment of a rational choice for
lation, or at least slow down the crystallisation rate and extent. the optimal amorphous system. The miscibility of the drug
In this context, the question of how much crystalline drug in with a polymer, the solubility in various solvents, the possibil-
an overall amorphous dosage form can be tolerated without ity for molecular interactions with various excipients, the ten-
compromising the bioavailability of the formulation needs dency for surface crystallisation, the melting temperature and
to be further investigated. This can be facilitated by the use thus the available production processes are just some examples
of physiologically relevant in vitro dissolution studies and ani- for variables that all influence the applicable processes and the
mal studies of amorphous dosage forms. Furthermore, the use final quality of the amorphous formulation. However, so far a
of analytical techniques that can clearly separate the total comprehensive understanding of the interactions between all
amount of crystallinity from the spatial distribution of the these variables is far from being achieved but this will be
crystalline and amorphous material is highly desirable and necessary in order to establish a rational basis for the optimal
requires the use of several orthogonal analytical techniques, choice of an amorphous formulation.
including the use of imaging techniques.
Salt formation is usually the initial step in improving the
dissolution properties of a drug and thus, rarely investigated Declaration of interest
for amorphous systems. However, the formation of an amor-
phous salt, for example between a basic amino acid and a The authors have no relevant affiliations or financial involve-
poorly soluble acidic drug, might even further improve the ment with any organisation or entity with a financial interest
dissolution rate in comparison to a crystalline salt. Prelimi- in or financial conflict with the subject matter or materials
nary studies in our group indicate that such co-amorphous discussed in the manuscript. This includes employment,
salts can remain amorphous, even in a slow evaporation consultancies, honoraria, stock ownership or options, expert
process, and thus might be unable to crystallise. testimony, grants or patents received or pending, or royalties.

Expert Opin. Drug Deliv. (2014) 11(6) 985


H. Grohganz et al.

Bibliography
Papers of special note have been highlighted as hydroxypropylmethylcellulose acetate 21. Huang JJ, Li Y, Wigent RJ, et al.
either of interest () or of considerable interest succinate (HPMCAS) for initiation and Interplay of formulation and process
() to readers. maintenance of drug supersaturation in methodology on the extent of nifedipine
the GI milieu. Pharm Res molecular dispersion in polymers.
1. Amidon GL, Lennernas H, Shah VP,
2009;26(6):1419-31 Int J Pharm 2011;420(1):59-67
et al. A theoretical basis for a
biopharmaceutic drug classification - the 12. Janssens S, Van den Mooter G. Review: 22. Tian YW, Booth J, Meehan E, et al.
correlation of in-vitro drug product physical chemistry of solid dispersions. Construction of drug-polymer
dissolution and in-vivo bioavailability. J Pharm Pharmacol thermodynamic phase diagrams using
Pharm Res 1995;12(3):413-20 2009;61(12):1571-86 Flory-Huggins interaction theory:
13. Linn M, Collnot EM, Djuric D, et al. identifying the relevance of temperature
2. Nair AK, Anand O, Chun N, et al.
Soluplus (R) as an effective absorption and drug weight fraction to phase
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

Statistics on BCS classification of generic


enhancer of poorly soluble drugs in vitro separation within solid dispersions.
drug products approved between
and in vivo. Eur J Pharm Sci Mol Pharm 2013;10(1):236-48
2000 and 2011 in the USA. AAPS J
2012;14(4):664-6 2012;45(3):336-43 23. Painter PC, Graf JF, Coleman MM.
14. Vasanthavada M, Tong WQ, Joshi Y, Effect of hydrogen-bonding on the
3. Babu NJ, Nangia A. Solubility advantage
et al. Phase behavior of amorphous enthalpy of mixing and the composition
of amorphous drugs and pharmaceutical
molecular dispersions I: determination of dependence of the glass-transition
cocrystals. Cryst Growth Des
the degree and mechanism of solid temperature in polymer blends.
2011;11(7):2662-79
solubility. Pharm Res Macromolecules 1991;24(20):5630-8
4. Martinez MN, Amidon GL.
2004;21(9):1598-606 24. Wertheim MS. Fluids with highly
A mechanistic approach to understanding
15. Mahieu A, Willart JF, Dudognon E, directional attractive forces. 1. Statistical
the factors affecting drug absorption:
et al. A new protocol to determine the thermodynamics. J Stat Phys
a review of fundamentals.
solubility of drugs into polymer matrices. 1984;35(1-2):19-34
J Clin Pharmacol 2002;42(6):620-43
Mol Pharm 2013;10(2):560-6 25. Al-Obaidi H, Brocchini S, Buckton G.
5. Hancock BC, Zografi G. Characteristics
For personal use only.

16. Sun Y, Tao J, Zhang GGZ, et al. Anomalous properties of spray dried
and significance of the amorphous state
Solubilities of crystalline drugs in solid dispersions. J Pharm Sci
in pharmaceutical systems. J Pharm Sci
polymers: an improved analytical method 2009;98(12):4724-37
1997;86(1):1-12
and comparison of solubilities of 26. Paudel A, Van den Mooter G. Influence
6. Hancock B, Parks M. What is the true
indomethacin and nifedipine in PVP, of solvent composition on the miscibility
solubility advantage for amorphous
PVP/VA, and PVAc. J Pharm Sci and physical stability of naproxen/PVP K
pharmaceuticals? Pharm Res
2010;99(9):4023-31 25 solid dispersions prepared by
2000;17(4):397-404
17. Marsac PJ, Shamblin SL, Taylor LS. cosolvent spray-drying. Pharm Res
7. Aaltonen J, Rades T. Commentary: 2012;29(1):251-70
Theoretical and practical approaches for
towards physicorelevant dissolution
prediction of drug-polymer miscibility 27. Kolasinac N, Kachrimanis K, Djuris J,
testing: the importance of solid-state
and solubility. Pharm Res et al. Spray coating as a powerful
analysis in dissolution.
2006;23(10):2417-26 technique in preparation of solid
Dissolution Technol 2009;16:47-54
18. Janssens S, De Zeure A, Paudel A, et al. dispersions with enhanced desloratadine
8. Grohganz H, Lobmann K, Priemel PA, dissolution rate. Drug Dev Ind Pharm
Influence of preparation methods on
et al. Amorphous drugs and dosage 2013;39(7):1020-7
solid state supersaturation of amorphous
forms. J Drug Deliv Sci Tec
solid dispersions: a case study with 28. Passerini N, Perissutti B, Moneghini M,
2013;23(4):403-8
itraconazole and eudragit E100. et al. Characterization of carbamazepine-
9. Chiou WL, Riegelman S. Pharmaceutical Pharm Res 2010;27(5):775-85 gelucire 50/13 microparticles prepared by
applications of solid dispersion systems. a spray-congealing process using
19. Paudel A, Van Humbeeck J,
J Pharm Sci 1971;60(9):1281-302 ultrasounds. J Pharm Sci
Van den Mooter G. Theoretical and
10. Alam MA, Ali R, Al-Jenoobi FI, et al. experimental investigation on the solid 2002;91(3):699-707
Solid dispersions: a strategy for poorly solubility and miscibility of naproxen in 29. Breitenbach J. Melt extrusion: from
aqueous soluble drugs and technology poly(vinylpyrrolidone). Mol Pharm process to drug delivery technology.
updates. Expert Opin Drug Deliv 2010;7(4):1133-48 Eur J Pharm Biopharm
2012;9(11):1419-40 . Different methods of calculation of the 2002;54(2):107-17
.. Recent review with relevant drug--polymer solubility are compared 30. DiNunzio JC, Brough C, Miller DA,
information, concerning materials, and discussed. et al. Fusion processing of itraconazole
methods of preparation and many
20. Bellantone RA, Patel P, Sandhu H, et al. solid dispersions by kinetisol (R)
examples. A good review to start
A method to predict the equilibrium dispersing: a comparative study to hot
learning this topic.
solubility of drugs in solid polymers near melt extrusion. J Pharm Sci
11. Curatolo W, Nightingale JA, room temperature using thermal analysis. 2010;99(3):1239-53
Herbig SM. Utility of J Pharm Sci 2012;101(12):4549-58

986 Expert Opin. Drug Deliv. (2014) 11(6)


Refining stability and dissolution rate of amorphous drug formulations

31. De Zordi N, Moneghini M, Kikic I, amorphous indomethacin. Eur J delivery of therapeutics. Expert Opin
et al. Applications of supercritical fluids Pharm Biopharm 2012;82(1):187-93 Drug Deliv 2007;4(2):101-10
to enhance the dissolution behaviors of 41. Bruce C, Fegely KA, 53. Shen S-C, Ng WK, Chia L, et al.
Furosemide by generation of Rajabi-Siahboomi AR, et al. Crystal Physical state and dissolution of
microparticles and solid dispersions. growth formation in melt extrudates. ibuprofen formulated by co-spray drying
Eur J Pharm Biopharm Int J Pharm 2007;341(1-2):162-72 with mesoporous silica: effect of pore
2012;81(1):131-41 and particle size. Int J Pharm
42. Wu T, Sun Y, Li N, et al. Inhibiting
32. Moneghini M, Kikic I, Voinovich D, surface crystallization of amorphous 2011;410(1 -- 2):188-95
et al. Processing of carbamazepine PEG indomethacin by nanocoating. Langmuir 54. Limnell T, Heikkila T, Santos HA, et al.
4000 solid dispersions with supercritical 2007;23(9):5148-53 Physicochemical stability of high
carbon dioxide: preparation, . Article highlighting the importance of indomethacin payload ordered
characterisation, and in vitro dissolution. coating on increasing stability of mesoporous silica MCM-41 and SBA-15
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

Int J Pharm 2001;222(1):129-38 amorphous particles. microparticles. Int J Pharm


33. Verreck G, Decorte A, Heymans K, et al. 43. Zhu L, Jona J, Nagapudi K, et al. Fast 2011;416(1):242-51
The effect of pressurized carbon dioxide surface crystallization of amorphous 55. Qian KK, Suib SL, Bogner RH.
as a temporary plasticizer and foaming griseofulvin below T g. Pharm Res Spontaneous crystalline-to-amorphous
agent on the hot stage extrusion process 2010;27(8):1558-67 phase transformation of organic or
and extrudate properties of solid medicinal compounds in the presence of
44. Zhu L, Wong L, Yu L. Surface-enhanced
dispersions of itraconazole with PVP-VA porous media, part 2: amorphization
crystallization of amorphous nifedipine.
64. Eur J Pharm Sci capacity and mechanisms of interaction.
Mol Pharm 2008;5(6):921-6
2005;26(3-4):349-58 J Pharm Sci 2011;100(11):4674-86
45. Priemel PA, Laitinen R, Barthold S,
34. Verreck G, Decorte A, Heymans K, et al. 56. Mellaerts R, Jammaer JAG,
et al. Inhibition of surface crystallisation
Hot stage extrusion of p-amino salicylic Van Speybroeck M, et al. Physical state
of amorphous indomethacin particles in
acid with EC using CO2 as a temporary of poorly water soluble therapeutic
physical drug-polymer mixtures.
plasticizer. Int J Pharm molecules loaded into SBA-15 ordered
Int J Pharm 2013;456(2):301-6
2006;327(1-2):45-50 mesoporous silica carriers: a case study
For personal use only.

46. Sun Y, Zhu L, Kearns KL, et al. Glasses


35. Nielsen LH, Keller SS, Gordon KC, with itraconazole and ibuprofen.
crystallize rapidly at free surfaces by
et al. Spatial confinement can lead to Langmuir 2008;24(16):8651-9
growing crystals upward. Proc Natl Acad
increased stability of amorphous 57. Wang F, Hui H, Barnes TJ, et al.
Sci USA 2011;108(15):5990-5
indomethacin. Eur J Pharm Biopharm Oxidized mesoporous silicon
2012;81(2):418-25 47. Bruce CD, Fegely KA,
microparticles for improved oral delivery
Rajabi-Siahboomi AR, et al. Aqueous
36. Nielsen LH, Keller SS, Boisen A, et al. of poorly soluble drugs. Mol Pharm
film coating to reduce recrystallization of
A slow cooling rate of indomethacin melt 2009;7(1):227-36
guaifenesin from hot-melt extruded
spatially confined in microcontainers 58. Ambrogi V, Perioli L, Pagano C, et al.
acrylic matrices. Film coating improves
increases the physical stability of the Use of SBA-15 for furosemide oral
physical stability of melt extruded tablets.
amorphous drug without influencing its delivery enhancement. Eur J Pharm Sci
Drug Dev Ind Pharm 2010;36(2):218-26
biorelevant dissolution behaviour. 2012;46(1 -- 2):43-8
Drug Deliv Transl Res 48. Ng YC, Yang Z, McAuley WJ, et al.
Stabilisation of amorphous drugs under 59. Chieng N, Aaltonen J, Saville D, et al.
2013;published online 14 August 2013;
high humidity using pharmaceutical thin Physical characterization and stability of
doi: 10.1007/s13346-013-0166-7
films. Eur J Pharm Biopharm amorphous indomethacin and ranitidine
37. Luth H. Solid surfaces, interfaces and hydrochloride binary systems prepared by
2013;84(3):555-65
thin films. 5th edition. Heidelberg mechanical activation. Eur J
Springer; New York: 2010 49. Rumondor ACF, Marsac PJ,
Pharm Biopharm 2009;71(1):47-54
Stanford LA, et al. Phase behavior of
38. Sun Y, Zhu L, Wu T, et al. Stability of 60. Alleso M, Chieng N, Rehder S, et al.
poly(vinylpyrrolidone) containing
amorphous pharmaceutical solids: crystal Enhanced dissolution rate and
amorphous solid dispersions in the
growth mechanisms and effect of synchronized release of drugs in binary
presence of moisture. Mol Pharm
polymer additives. AAPS J systems through formulation: amorphous
2009;6(5):1492-505
2012;14(3):380-8 naproxen-cimetidine mixtures prepared
50. Xu W, Riikonen J, Lehto V-P.
39. Weber R, Grotkopp I, Stettner J, et al. by mechanical activation.
Mesoporous systems for poorly soluble
Embedding of gold nanoclusters on J Control Release 2009;136(1):45-53
drugs. Int J Pharm 2013;453(1):181-97 .
polystyrene surfaces: influence of the Highlights possibilities of co-
surface modification on the glass 51. Qian KK, Bogner RH. Application of amorphpous drug--drug combinations
transition. Macromolecules mesoporous silicon dioxide and silicate in to increase and synchronise the release.
2003;36(24):9100-6 oral amorphous drug delivery systems.
61. Lobmann K, Laitinen R, Grohganz H,
J Pharm Sci 2012;101(2):444-63
40. Priemel PA, Grohganz H, Gordon KC, et al. Coamorphous drug systems:
et al. The impact of surface- and nano- 52. Prestidge CA, Barnes TJ, Lau C-H, et al. enhanced physical stability and
crystallisation on the detected amorphous Mesoporous silicon: a platform for the dissolution rate of indomethacin and
content and the dissolution behaviour of

Expert Opin. Drug Deliv. (2014) 11(6) 987


H. Grohganz et al.

naproxen. Mol Pharm 72. Hussain MD, Saxena V, Brausch JF, furosemide bioavailability:
2011;8(5):1919-28 et al. Ibuprofen--phospholipid solid a bioequivalence study. Pharm Res
62. Lobmann K, Strachan C, Grohganz H, dispersions: improved dissolution and 1987;4(2):150-3
et al. Co-amorphous simvastatin and gastric tolerance. Int J Pharm 83. Matsuda Y, Tatsumi E. Physiochemical
glipizide combinations show improved 2012;422(1 -- 2):290-4 characterization of furosemide
physical stability without evidence of 73. Ahuja N, Katare OP, Singh B. Studies modifications. Int J Pharm 1990;60:16
intermolecular interactions. Eur J on dissolution enhancement and 84. Nielsen LH, Gordon S, Holm R, et al.
Pharm Biopharm 2012;81(1):159-69 mathematical modeling of drug release of Preparation of an amorphous sodium
63. Lobmann K, Laitinen R, Grohganz H, a poorly water-soluble drug using water- furosemide salt improves solubility and
et al. A theoretical and spectroscopic soluble carriers. Eur J Pharm Biopharm dissolution rate and leads to a faster T
study of co-amorphous naproxen and 2007;65(1):26-38 after oral dosing to rats. Eur J
indomethacin. Int J Pharm 74. Masuda T, Yoshihashi Y, Yonemochi E, Pharm Biopharm 2013;85(3B):942-51
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

2013;453(1):80-7 et al. Cocrystallization and amorphization 85. Nielsen LH, Gordon S, Pajander JP,
64. Lobmann K, Grohganz H, Laitinen R, induced by drug--excipient interaction et al. Biorelevant characterisation of
et al. Amino acids as co-amorphous improves the physical properties of amorphous furosemide salt exhibits
stabilizers for poorly water soluble drugs acyclovir. Int J Pharm conversion to a furosemide hydrate
-- Part 1: preparation, stability and 2012;422(1 -- 2):160-9 during dissolution. Int J Pharm
dissolution enhancement. Eur J 75. Hoppu P, Jouppila K, Rantanen J, et al. 2013;457(1):14-24
Pharm Biopharm 2013;85(3B):873-81 Characterisation of blends of paracetamol 86. Nielsen LH, Rades T, Mullertz A.
. First example of co-amorphous and citric acid. J Pharm Pharmacol Characterisation during storage and
drug--amino acid combinations. 2007;59(3):373-81 dissolution of solid dispersions
65. Lobmann K, Laitinen R, Strachan C, 76. Corrigan OI, Holohan EM, Sabra K. containing furosemide and
et al. Amino acids as co-amorphous Amorphous forms of thiazide diuretics hydroxypropyl methylcellulose. J Drug
stabilizers for poorly water-soluble drugs prepared by spray-drying. Int J Pharm Deliv Sci Tech 2013;23(4):409-15
-- Part 2: molecular interactions. Eur J 1984;18:6 87. Priemel PA, Laitinen R, Grohganz H,
Pharm Biopharm 2013;85(3B):882-8
For personal use only.

77. Paluch KJ, Tajber L, Corrigan OI, et al. et al. In situ amorphisation of
66. Yu L. Amorphous pharmaceutical solids: Impact of process variables on the indomethacin with Eudragit E during
preparation, characterization and micromeritic and physicochemical dissolution. Eur J Pharm Biopharm
stabilization. Adv Drug Deliv Rev properties of spray-dried porous 2013;85(3B):1259-65
2001;48(1):27-42 microparticles, part I: introduction of a . Article highlighting the potential of in
67. Lobmann K, Jensen K, Laitinen R, et al. new morphology classification system. situ amorphisation prior
Formulating poorly water soluble drugs J Pharm Pharmacol to administration.
into co-amorphous drug-amino acid 2012;64(11):1570-82 88. Karmwar P, Graeser K, Gordon KC,
mixtures. In: Qu H, Rantanen J, 78. Sonje VM, Kumar L, Puri V, et al. et al. Effect of different preparation
Malwade C, editors, BIWIC 2013: 20th Effect of counterions on the properties of methods on the dissolution behaviour of
international workshop on industrial amorphous atorvastatin salts. Eur J amorphous indomethacin. Eur J
crystallization. University of Southern Pharm Sci 2011;44(4):462-70 Pharm Biopharm 2012;80(2):459-64
Denmark; Odense, Denmark: 79. Bianco S, Caron V, Tajber L, et al. 89. Mahmah O, Tabbakh R, Kelly A, et al.
2013. p. 31-7 Modification of the solid-state nature of A comparative study of the effect of
68. Lu Q, Zografi G. Phase behavior of sulfathiazole and sulfathiazole sodium by spray drying and hot-melt extrusion on
binary and ternary amorphous mixtures spray drying. AAPS PharmSciTech the properties of amorphous solid
containing indomethacin, citric acid, and 2012;13(2):647-60 dispersions containing felodipine.
PVP. Pharm Res 15(8):1202-6 80. Paluch KJ, Tajber L, Amaro MI, et al. J Pharm Pharmacol 2014;66(2):275-84
69. Hu Y, Gniado K, Erxleben A, et al. Impact of process variables on the 90. Patterson JE, James MB, Forster AH,
Mechanochemical reaction of micromeritic and physicochemical et al. Preparation of glass solutions of
sulfathiazole with carboxylic acids: properties of spray-dried microparticles -- three poorly water soluble drugs by spray
formation of a cocrystal, a salt, and Part II. Physicochemical characterisation drying, melt extrusion and ball milling.
coamorphous solids. Cryst Growth Des of spray-dried materials. Int J Pharm 2007;336(1):22-34
2013;14(2):803-13 J Pharm Pharmacol 91. Ozaki S, Kushida I, Yamashita T, et al.
70. Telang C, Mujumdar S, Mathew M. 2012;64(11):1583-91 Evaluation of drug supersaturation by
Improved physical stability of amorphous 81. Bianco S, Tewes F, Tajber L, et al. Bulk, thermodynamic and kinetic approaches
state through acid base interactions. surface properties and water uptake for the prediction of oral absorbability in
J Pharm Sci 2009;98(6):2149-59 mechanisms of salt/acid amorphous amorphous pharmaceuticals. J Pharm Sci
71. Gao Y, Liao J, Qi X, Zhang J. composite systems. Int J Pharm 2012;101(11):4220-30
Coamorphous repaglinide--saccharin with 2013;456(1):143-52 92. Warren DB, Bergstrom CAS,
enhanced dissolution. Int J Pharm 82. McNamara PJ, Foster TS, Digenis GA, Benameur H, et al. Evaluation of the
2013;450(1 -- 2):290-5 et al. Influence of tablet dissolution on structural determinants of polymeric

988 Expert Opin. Drug Deliv. (2014) 11(6)


Refining stability and dissolution rate of amorphous drug formulations

precipitation inhibitors using solvent shift after administration of two weak bases to Affiliation
methods and principle component fasted adults. Pharm Res Holger Grohganz1, Petra A Priemel1,2,
analysis. Mol Pharm 2013;10(8):2823-48 2011;28(12):3145-58 Korbinian Lobmann1, Line Hagner Nielsen1,
93. Wyttenbach N, Janas C, Siam M, et al. 97. Sassene PJ, Knopp MM, Hesselkilde JZ, Riikka Laitinen3, Anette Mullertz1,
Miniaturized screening of polymers for et al. Precipitation of a poorly soluble Guy Van den Mooter4 & Thomas Rades1

amorphous drug stabilization (SPADS): model drug during in vitro lipolysis: Author for correspondence
1
rapid assessment of solid dispersion characterization and dissolution of the University of Copenhagen, Department of
systems. Eur J Pharm Biopharm precipitate. J Pharm Sci Pharmacy, Universitetsparken 2,
2013;84(3):583-98 2010;99(12):4982-91 2100 Copenhagen, Denmark
94. Bevernage J, Brouwers J, Annaert P,
. Article highlighting the potential of in Tel: +45 3533 6032;
et al. Drug precipitation--permeation situ amorphisation during in E-mail: Thomas.rades@sund.ku.dk
2
interplay: supersaturation in an vitro lipolysis. University of Otago, School of Pharmacy,
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Nanyang Technological University on 06/04/14

Dunedin, New Zealand


absorptive environment. Eur J 98. Thomas N, Holm R, Mullertz A, et al. 3
University of Eastern Finland, School of
Pharm Biopharm 2012;82(2):424-8 In vitro and in vivo performance of
Pharmacy, Kuopio, Finland
95. Carlert S, Akesson P, Jerndal G, et al. In novel supersaturated self-nanoemulsifying 4
University of Leuven, Department of
vivo dog intestinal precipitation of drug delivery systems (super-SNEDDS).
Pharmaceutical and Pharmacological Sciences,
mebendazole: a basic BCS class II drug. J Control Release 2012;160(1):25-32
Leuven, Belgium
Mol Pharm 2012;9(10):2903-11 99. Thomas N, Rades T, Mullertz A. Recent
96. Psachoulias D, Vertzoni M, Goumas K, developments in oral lipid-based drug
et al. Precipitation in and supersaturation delivery. J Drug Deliv Sci Tech
of contents of the upper small intestine 2013;23(4):375-82
For personal use only.

Expert Opin. Drug Deliv. (2014) 11(6) 989

Anda mungkin juga menyukai