Anda di halaman 1dari 15

PHARMACEUTICAL TECHNOLOGY

Physicochemical Properties of the Amorphous Drug, Cast Films,


and Spray Dried Powders to Predict Formulation Probability of
Success for Solid Dispersions: Etravirine
ILSE WEUTS,1 FREDERIC VAN DYCKE,1 JODY VOORSPOELS,1 STEVE DE CORT,1 SIGRID STOKBROEKX,1 RUUD LEEMANS,1
MARCUS E. BREWSTER,1 DAWEI XU,2 BRIGITTE SEGMULLER,2 YA TSZ A. TURNER,3 CLIVE J. ROBERTS,3
MARTYN C. DAVIES,3 SHENG QI,4 DUNCAN Q.M. CRAIG,4 MIKE READING4
1
Chemical and Pharmaceutical Development, Johnson & Johnson, Beerse, Belgium
2
Analytical Development, Johnson & Johnson Pharmaceutical R&D, LLC, Raritan, New Jersey 08869
3
Laboratory of Biophysics and Surface Analysis, School of Pharmacy, University of Nottingham, University Park, Nottingham,
NG7 2RD, United Kingdom
4
School of Chemical Sciences and Pharmacy, University of East Anglia, Norwich, United Kingdom

Received 31 January 2010; revised 23 April 2010; accepted 26 April 2010


Published online 22 June 2010 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.22242

ABSTRACT: Solid dispersion technology represents an enabling approach to formulate poorly


water-soluble drugs. While providing for a potentially increased oral bioavailability secondary to
an increased drug dissolution rate, amorphous dispersions can be limited by their physical
stability. The ability to assess formulation risk in this regard early in development programs can
not only help in guiding development strategies but can also point to critical design elements in
the configuration of the dosage form. Based on experience with a recently approved solid
dispersion-based product, Intelence1 (etravirine), a three part strategy is suggested to predict
early formulate-ability of these systems. The components include an assessment of the amor-
phous form, a study of binary drug/carrier cast films and the evaluation of a powder of the drug
and polymer processed in a manner relevant to the intended final dosage form. A variety of
thermoanalytical, spectroscopic, and spectrophotometric approaches were applied to study the
prepared materials. The data suggest a correlation between the glass forming ability and
stability of the amorphous drug and the nature of the final formulation. Cast films can provide
early information on miscibility and stabilization and assessment of processed powders can help
define requirements and identify issues with potential final formulations. 2010 Wiley-Liss, Inc.
and the American Pharmacists Association J Pharm Sci 100:260274, 2011
Keywords: solid dispersion; characterization; amorphous; etravirine; HPMC

INTRODUCTION in contemporary drug pipelines are thought to


contribute to this overall decrease in drugability
The development of new pharmaceutical agents including the manner in which drugs are discovered
is often confounded by their poor water solubility and the chemically allowed space of the drug target.
which impacts a number of derivative properties As reviewed by Lipinski,1,3 high throughput screen-
including poor dissolution rate.1,2 Inadequate solu- ing has tended to select for compounds with lower
bility or dissolution rate can significantly reduce both water-solubilities, higher lipophilicities and higher
the rate and extent of drug absorption deleteriously molecular weights. Furthermore, many drug targets
affecting oral bioavailability. Several factors rooted have structure-activity relationship (SAR) require-
ments that do not overlap with properties known to
provide for good oral bioavailability resulting in poor
Correspondence to: Ilse Weuts (Telephone: 32-14-608205;
Fax: 32-14-607083; E-mail: iweuts@its.jnj.com) develop-ability and high attrition.4,5 While expanding
Journal of Pharmaceutical Sciences, Vol. 100, 260274 (2011) the chemical space of the target is the purview of
2010 Wiley-Liss, Inc. and the American Pharmacists Association medicinal chemistry, pharmaceutical scientists can

260 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011


PREDICTION OF FORMULATION PROBABILITY OF SUCCESS FOR SOLID DISPERSIONS 261

contribute to enlarging the oral drug bioavailability Table 1. Physicochemical and Solubility Properties of
space through the use of technologies that increase Etravirine
apparent solubility and dissolution rate. A number
Property Value
of approaches are available for increasing the
formulate-ability of BCS Class II compounds includ- Molecular weight (g/mol) 435
ing but not limited to complexation, micronization or Molecular formula C20H15BrN6O
Melting point 2608C (dec.)
nanonization, use of cosolvents and structured lipid
log p >5
systems.68 Solid amorphous dispersions provide a pKa (base) <3
unique opportunity in this regard.911 While theore- S (water) 1 mg/mL
tically of benefit in any number of situations, the S (0.1 N HCl) 1 mg/mL
physical instability of solid amorphous dispersions S (PEG400) 74 mg/mL
often conspires to limit their usefulness. Early
assessments of factors likely to impact on the
applicability of a dispersion based approach such as of powders prepared using scaled-down processes
stability, are important components in any develop- simulating those likely to apply to the finished dosage
ment program centered around such a formulation form. Thus, this communication examines the proper-
concept. We suggest herein a three pronged approach ties of amorphous etravirine,16 the characteristics of
for assessing dispersion fitness using the recently solvent cast films containing the drug and HPMC and
approved drug, Intelence1 (etravirine) as an illus- assessments of the physical state of the drug and its
trative example. miscibility with the polymer as a function of drug/
Etravirine (Fig. 1) is a novel second generation non- polymer ratio in spray dried powders.15,1720 These
nucleoside reverse transcriptase inhibitor (NNRTI) assessments required a number of appropriate
which acts by molecularly blocking the viral reverse and complementary analytical methods such as
transcriptase enzyme. It achieves this by preventing powder X-ray diffraction (XRD), infrared spectro-
the enzyme from converting its genetic material scopy (IR), differential scanning calorimetry (DSC),
(RNA) into proviral DNA and thus preventing modulated temperature differential scanning calori-
incorporation of the viral genome into the human metry (M-DSC) and solid state nuclear magnetic
host cell.12,13 The physicochemical properties of resonance (SS-NMR). Imaging techniques such as
etravirine are challenging with regard to dosage field emission gun-environmental scanning electron
form configuration as indicated in Table 1. A variety microscopy (FEG-ESEM) and atomic force micro-
of technologies were screened to optimize bioavail- scopy (AFM)2123 allowed the spatial assessment of
ability, however most did not add significant value. pharmaceutical formulations with resolution up to
For example, oral dosing of a nanosuspension the nanometer scale.
resulted in negligible blood levels in dog. The best
enhancement of GI uptake was provided by systems
EXPERIMENTAL
based on an amorphous drug substance and specifi-
cally, amorphous dispersions of the compound in a
Materials
glassy carrier.911,14,15
These initial screening elements were the inputs Etravirine (Fig. 1) was obtained from Janssen
for a development program to fabricate a viable Pharmaceutica (Beerse, Belgium). The hydroxypro-
formulation for etravirine. In devising this program, pylmethyl cellulose (HPMC) was 2910 grade with a
factors thought to contribute to, or detract from viscosity of 5 mPa s and was purchased from Dow
success were posited. The philosophy adopted Chemical (Plaquemine, LA). Amorphous etravirine
included (1) a study of the pure amorphous drug was prepared by cryomilling the crystalline drug
phase, (2) an evaluation of cast films of the drug and a substance for 3 h at 1968C. Other methods such
glassy polymer (HPMC), and (3) an assessment as spray drying, freeze drying, and ball milling
were unsuccessful in generating stable amorphous
etravirine (data not shown). Stability was assessed
using FT-IR as described in the literature.16
Film Casting
A screening study found two useful solvent mixtures
including dichloromethane (DCM)-ethanol (80:20)
and various compositions of dimethylformamide
(DMF)-methanol (80:20, 50:50, and 40:60). Drug
and polymer (HPMC) were dissolved in the solvent
Figure 1. Chemical structure of etravirine. mixture of interest and the solutions were filtered and

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011
262 WEUTS ET AL.

cast onto a Teflon plate (15 cm  15 cm). The solvent preparation of calibration curves, a sample holder
was evaporated in a vacuum oven at between 85 and with a cavity of 16 mm diameter was implemented.
1258C for several days until a constant weight was This methodology provided for improved robustness,
obtained. Films were then subjected to M-DSC for increased sensitivity and was less susceptible to
determination of Tg and the enthalpy of relaxation as preferred orientation effects than the zero back-
well as XRD for assessing crystallinity. ground holder. Bragg-Brentano geometry was
applied using monochromatic Cu Ka-radiation and
Spray-Dried Powders
soller slits (0.04 rad) for the incident and diffracted
Solid dispersions of various compositions were prepared beam path. The instrument was operated at a voltage
by spray drying on a laboratory scale instrument of 45 kV and a current of 40 mA. The scan range was
(NIRO atomizer mobile minor, Soeberg, Denmark). from 3 to 508 2u with a step size of 0.028 and a counting
Etravirine and HPMC were dissolved in an organic time of 60 or 100 s per step. Crystallinity was
solvent mixture (dichloromethane (DCM)/ethanol), estimated by constructing a standard curve in which
after which the solvent was removed by spray drying. spray-dried powders of HPMC and crystalline etra-
A dual fluid nozzle with a diameter of 1 mm was virine were mixed with amorphous powders providing
applied. Solutions were sprayed using a feed rate of a range from 0% to 100% crystalline content. The
34 kg/h, an atomization pressure of 0.8 bars, an inlet peak area associated with the diffraction peak
temperature of 1108C and an outlet temperature of between 8 and 108 2u (see Fig. 3b) was assessed as
708C. These parameters were maintained for the a function of the percent crystalline content. There
different dispersions. However, the solvent composi- was a good linear response (r  0.99) and low
tion and the concentration of etravirine and HPMC variability (samples of the calibration curve were
in the feed solutions varied (generating changes in measured in triplicate) at crystalline contents up to
solvent evaporation rate and solution viscosity). approximately 30% or 40%. At higher crystalline
Powders with the following etravirine/HPMC ratios compositions, the linear fit of the data decreased and
(w/w) were prepared: 1:12, 1:9, 1:6, 1:5, 1:3, 1:2, 1:1, variability increased. The limit of detection was
1:0.5, 1:0.3, 1:0.1, and 0:1. approximately 2% and the lower limit of quantitation
Physical mixtures were prepared by gently mixing was approximately 6%.
amorphous, cryomilled etravirine and spray dried
Fourier-Transform Infra-Red Spectroscopy (FT-IR)
HPMC using a pestle and mortar. Powders with
the following etravirine/HPMC ratios (w/w) were FT-IR spectra were measured using a Nicolet Nexus
prepared: 1:3, 1:1, and 1:0.5. 670 FT-IR spectrometer (Thermo Fisher Scientific,
Waltham, MA) equipped with micro attenuated total
Modulated Temperature Differential Scanning
reflectance (microATR). Thirty two scans were taken
Calorimetry (M-DSC)
for each sample in the spectral range from 400 to
The cast films or powder samples were analyzed using 4000 cm1 with a spectral resolution of 1 cm1.
a TGA Q-1000 MDSC (TA Instruments, Crawley, UK)
Fourier-Transform Raman Spectroscopy (FT-Raman)
with a heating rate of 28C/min, an amplitude of
0.3188C and a period of 60 s. Standard aluminum DSC The powders were transferred to a glass capillary cell
pans (TA Instruments) were used without crimping. and spectra were recorded on a Nicolet FT-Raman
module (Thermo Fisher Scientific). 128 or 256 scans
Differential Scanning Calorimetry (DSC)
were taken for each sample in the spectral range
The assessment of the Tg of the amorphous etravirine from 100 to 4000 cm1 with a spectral resolution of
could most easily be performed using thermal 4 cm1.
analysis without temperature modulation. The pow-
Solid State-Nuclear Magnetic Resonance
der was heated in the same pan type as for the M-DSC
Spectroscopy (SS-NMR)
measurements but higher heating rates were applied
(from 10 up to 1008C/min). The number of samples that were analyzed using this
technique was limited to the following: pure compo-
Powder X-Ray Diffraction Analysis (P-XRD)
nents (Etravirine (both amorphous and crystalline)
P-XRD-analyses were performed on a Philips XPert and HPMC), solid dispersions (ratios 1:3, 1:1, and
PRO MPD diffractometer PW3050/60 with a PW3040 1:0.5) and physical mixtures of the same ratios.
generator (PANalytical, Almelo, The Netherlands). Solid-state carbon CP/MAS NMR data were collected
The instrument is equipped with a Cu LFF X-ray tube using a Bruker Avance 400 MHz widebore NMR
PW3373/10. The cast films or powders were applied to spectrometer equipped with a 4 mm MAS probe
a zero background holder and placed on a spinner (Bruker BioSpin Corporation, Billerica, MA), with
stage with a spinner revolution time of 1 s. For the Etravirine formulation samples spun at 5000 Hz
quantitation of crystalline content including the and a contact time of 3.5 ms at 258C. Measurements

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011 DOI 10.1002/jps
PREDICTION OF FORMULATION PROBABILITY OF SUCCESS FOR SOLID DISPERSIONS 263

of amorphous Etravirine and of its physical mixtures Raman-spectra clearly show a distinct spectrum for
with HPMC were performed at low temperature the forms of interest in which the amorphous material
(508C), due to the thermal instability of the was characterized by the absence of sharp absorption
amorphous state in these samples. Proton spin- bands which are typical for crystalline systems.
lattice relaxation times (T1 and T1r) were measured The FT-IR spectra of cryomilled etravirine and the
as well. crystalline polymorphic Forms I and II are shown in
Figure 2. Three regions where the different forms
Field Emission Gun Environmental Scanning Electron
demonstrate the greatest divergence in spectral
Microscopy (FEG-ESEM)
characteristics have been highlighted. Similarly,
The powders were sprinkled onto double sided carbon peak broadening is observed in SS-NMR-13C-spectra
attached to a standard SEM platform, and any excess (data not shown). XRD-analysis showed the absence
powder on the surface was removed by a stream of of discrete diffraction peaks. A halo, typical for
nitrogen. The uncoated samples were studied using materials that lack long range order, is observed.
a Philips FEI XL30 ESEM-FEG under a stream of The diffraction pattern of amorphous etravirine and
nitrogen gas. The vapor pressure was kept between two of its crystalline forms are shown in an overlay in
1.8 and 1.9 Torr to minimize interference. A low Figure 3a and b. M-DSC-analysis indicates a glass
accelerating voltage of 10 kV at a working distance of transition (Tg) at 998C however the Tg observed in
10 mm and a beam current of 34 nA were employed. the reversible heat flow signal is accompanied by
recrystallization of the drug (exothermic event in the
Atomic Force Microscopy (AFM)
irreversible and total heat flow signal). In order to
A small amount of powder was sprinkled on to the ensure that the phenomenon ascribed to the Tg
surface of a thin layer of araldite glue. After 10 min, observed in the reversible heat flow is a true Tg and
excess powder was removed under a gentle stream of not an artifact caused by a spurious deconvolution of
nitrogen. Topographic height and phase images were the total heat flow associated with the reversible and
simultaneously obtained using tapping mode AFM nonreversible components, nonmodulated DSC mea-
(TM-AFM) with a Nanoscope IIIa Multimode AFM surements were also performed. High heating rates
system equipped with either an E or J scanner (Veeco can inhibit kinetic phenomena such as cold crystal-
InstrumentsTM, Santa Barabra, CA) in air. NP tips on lization or delay them to higher temperatures.27
125 mm silicon cantilevers (Tap300, Budget SensorTM, Figure 4 shows the DSC-curves that were recorded
Sophia, Bulgaria) with a resonant frequency between at different heating rates. A Tg around 998C was
200 and 400 kHz were used. Scan rates between 1 and 2 Hz observed, supporting the assumption that the phe-
were used to help minimize tip-sample interactions. nomenon that was observed in the reversible heat
flow curve of the M-DSC, is the glass transition. The
anomalously high DCp at 1008C/min may be asso-
RESULTS
ciated with better differentiation of the glass transi-
tion and cold crystallization events which may
Amorphous Drug Substance
overlap at lower scanning rate.
The availability and study of pure amorphous The generated thermoanalytical data can shed
etravirine would add value to the characterization light on the stability of the amorphous phase (Tab. 2).
of solid dispersions by not only providing a standard The Tg/Tm ratio for a variety of pharmaceutical glass
to assess drugpolymer mixtures but also as a tool to formers falls in a range of 0.720.79 and can provide a
gauge the nature of the formulation challenge and general sense of glass fragility and the tendency to
suggest the best development options to address these crystallize.2833 This value is difficult to generate for
issues.2426 A number of approaches were attempted etravirine based on the fact that compound melting
to generate the amorphous material. Based on the occurs coincident with degradation. To estimate the
thermal lability some of these were not appropriate melting point, DSC experiments using high to very
for this compound such as those related to melting high scanning rates were employed. These data
and melt quenching. Spray-drying, ball milling, suggested a Tg/Tm of 0.69 which could suggest a
freeze-drying, rapid precipitation and rapid solvent fragile system.28 The stability of the generated form
evaporation (using a variety of conditions including was next assessed. Anecdotal information found that
flash evaporation using hyper-DSC) all provided for the amorphous material began to crystallize when
material containing significant crystalline content. stored in open conditions at ambient temperature
The amorphous material was ultimately prepared by in a few days. This was confirmed by a controlled
cryomilling (3 h at 1968C).16 The amorphous nature assessment using FT-IR to assess spectral changes as
of the material was confirmed by spectroscopic (FT- a function of time. Even when the material was stored
IR, FT-Raman, SS-NMR) and thermal techniques at 308C below its glass transition temperature,
(DSC, M-DSC) as well as powder XRD. The IR- and significant and rapid form conversion occurred.16

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011
264 WEUTS ET AL.

Figure 2. FT-IR spectra (from top to bottom): cryomilled etravirine, etravirine poly-
morph II, and polymorph I.

DrugHPMC Cast Films 1:3 was assessed (Tab. 3). As a function of annealing
temperature (i.e., storage temperature below Tg) and
Screening studies were performed to select an optimal time, the extent of relaxation varied consistent with
common solvent from which drugpolymer (HPMC) theory. Furthermore, the rank order suggested that
films could be cast.31 DCM-ethanol allowed for useful the higher the polymer content, the lower the extent
films but only to a drug loading of 20% w/w. Above this of relaxation and the greater the thermodynamic
drugpolymer ratio, a crystalline component in the stability. While devitrification appears to be disfa-
glass was evident upon casting. Improved results vored at higher polymer ratios, other kinetic factors
were found with DMF-methanol which allowed can drive drug crystallization.32 To assess these, XRD
amorphous films with up to a 70% w/w drug loading was applied to the cast films and the approximate
to be prepared. This finding was interesting in that extent of crystallization assessed (using a standard
even though the evaporation rate of this solvent curve). The conditions examined included storage
systems is likely to be lower than that of the DCM temperatures above the Tg such that crystallization
ethanol mixture (based on the differences in solvent disconnected from relaxation could be studied. Within
vapor pressures) which would be expected to aid drug the limitations of this method (see the Experimental
crystallization, the solubility of the components in Section), a higher polymer content resulted in a
the DMFmethanol solvent was higher, reducing the longer induction period when the films were stored
extent of supersaturation and the crystallization at Tg (Tab. 4) and on a lower crystalline content and
driving force. Films were therefore prepared using crystallization rate at temperature above their Tg.
this solvent at different drug and HPMC composi- These data point to the dramatic stabilizing effect of
tions.31 The Tg varied as a function of drugpolymer the polymer when the drug is made amorphous by
ratio suggesting that a higher polymer content film casting in HPMC in comparison to the cryomilled
increased Tg (drug/polymer ratio 1:1; Tg 1068C, (amorphous) API.
1:2 ratio; Tg 1118C and 1:3 ratio; Tg 1148C). The
Processed DrugHPMC Powders
cast films were then evaluated using M-DSC to assess
the enthalpies of relaxation. The extent of relaxation Drugpolymer powders prepared using the proces-
at three drug to polymer ratios namely 1:1, 1:2, and sing technique of interest were assessed. Powders

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011 DOI 10.1002/jps
PREDICTION OF FORMULATION PROBABILITY OF SUCCESS FOR SOLID DISPERSIONS 265

Figure 3. (a) Overlay of XRD-patterns of amorphous dispersions with different


etravirine/HPMC ratio (from top to bottom: etravirine/HPMC 1:3, 1:1, 1:0.5, amorphous
etravirine, spray-dried HPMC). (b) Overlay of XRD-patterns of partially amorphous
dispersions with different etravirine/HPMC ratio (from top to bottom: etravirine/HPMC
1:0.3, 1:0.1, etravirine polymorph II, polymorph I).

were prepared by spray-drying the drug at different amorphous. Importantly, these data do not exclude
ratios (etravirine/HPMC) specifically: 1:12, 1:9, 1:6, the presence of some crystalline etravirine in the
1:5, 1:3, 1:2, 1:1, 1:0.5, 1:0.3, 1:0.1, and 0:1 w/w (spray samples. In any case, the Tg was found to increase
dried HPMC). In addition, physical mixtures of the with decreasing drug/polymer ratio which implies
drug and HPMC were also generated at ratios of 1:3, that the composition of the amorphous phase is
1:1, and 1:0.5. Figure 5 shows an overlay of the dependent on the drug/polymer ratio. In Figure 6, the
reversing heat flow curves of etravirine/HPMC measured Tg-values are plotted as a function of drug
dispersions with varying drug/polymer ratio. All concentration. The Tg changes with composition
the dispersions exhibit a single Tg that falls in especially in polymer rich dispersions. In the drug-
between the Tgs of the two dispersed components rich dispersions (greater than 1:1 drug/polymer) Tgs
(998C for etravirine and 1478C for HPMC), which in the range of the pure amorphous phase were
indicates that all the dispersions are at least partially recorded and the changes in Tg as a function of

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011
266 WEUTS ET AL.

Figure 4. DSC thermograms assessing Tg at different heating rates for cryomilled


(amorphous) etravirine.

composition were less dramatic. Figure 6 also gives be approximated by the SimhaBoyer rule:34
the theoretical Tg-values calculated using the
GordonTaylor equation.33 This relationship can be r1 Tg1
K
used to estimate the Tg in binary mixtures; however, r2 Tg2
this relationship gives accurate predictions only in
the case of ideal mixing, that is, when two conditions where r1 and r2 are the densities of the API and
are being met: both components are mixed at the the polymer. The Tg values and true densities for
molecular level (no phase separation) and mixing is etravirine and HPMC are 373 K (Tab. 2), 1.439 g/cm3
not accompanied by volume changes, that is, homo- (density of crystalline etravirine) and 413 K and
and heteromolecular forces are of equal strength. The 1.326 g/cm3 for HPMC. The density of amorphous
GordonTaylor equation is given as: etravirine was estimated as 0.95 that of the crystal-
line density based on the literature, resulting in a
w1 Tg1 Kw2 Tg2 value of 1.515 g/cm3.35
Tgmix
w1 Kw2
Table 3. The Extent of Relaxation (%) as a Function of
where w1 and w2 are the weight fractions of etravirine Annealing Temperature and Storage Time in Cast Films of
and HPMC and Tg1 and Tg2 are their glass transition Etravirine and HPMC
temperatures. Tg,mix is the glass transition tempera-
ture of the solid dispersion. K is a constant which can Drug/Polymer Drug/Polymer Drug/Polymer
Time (h) 1:1 (%) 1:2 (%) 1:3 (%)

Tg 25 K
Table 2. Thermal Properties Related to Amorphous 8 44 26 3.2
Etravirine and Itraconazole 24 49 40 5
170 54 48 11
Property Etravirine Itraconazole4042 570 61 52 22
Tg 35 K
Melting point 537 Ka 440.3 K 8 31 12 1
Glass transition temp. (Tg) 373 K 332.4 K 24 37 20 2
Heat capacity (Cp) at Tg 0.23 J/g K 0.43 J/g K 170 40 23 4
Enthalpy of fusion (DHfus) 100 J/ga 84.5 J/g 570 55 30 6
Tm/Tg 0.69 0.76 Tg 45 K
Activation energy of structural 905 kJ/mol 8 7 1 0
relaxation at Tg 24 11 4 0
Fragility parameter (m) 103 170 26 6 1
a
570 30 10 2
Melts with decomposition.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011 DOI 10.1002/jps
PREDICTION OF FORMULATION PROBABILITY OF SUCCESS FOR SOLID DISPERSIONS 267

Table 4. The Extent of Crystallization (%) as a Function It is clear from Figure 6 that the experimental Tgs
of Temperature and Storage Time in Cast Films of deviate from the calculated values.
Etravirine and HPMC Figure 7 shows an overlay of the total heat
flow curves of the spray-dried dispersions with
Drug/Polymer Drug/Polymer Drug/Polymer
Time (h) 1:1 (%) 1:2 (%) 1:3 (%) different drug/polymer ratios. They illustrate that
once the temperature is above the Tg, etravirine
Tg 25 K crystallizes causing the observed exothermic event in
3 16 9 8
8 23 14 12
the DSC-curves. The data indicate that the barrier
24 30 22 18 to crystallization increases as the polymer load
96 31 30 30 increases, that is, crystallization takes place at higher
120 36 36 31 temperature.
144 38 36 31 Melting of etravirine takes place above 2008C
193 43 37 31
Tg 10 K
(temperature region not included in the graph)
3 4a <LOD <LOD and is accompanied by decomposition. Hence heats
8 7 4a <LOD of fusion had to be estimated by rapid scanning
24 12 7 5a techniques. As a consequence, the presence of
96 14 12 10 crystalline etravirine in the spray dried dispersions
120 23 14 10
144 24 15 12
cannot be accurately assessed by comparing the heat
193 25 16 12 of fusion in the dispersions to the crystallization
Tg enthalpy of the drug. The DSC-analyses, therefore,
3 <LOD <LOD <LOD suggest that the etravirine/HPMC materials consist
8 <LOD <LOD <LOD of one amorphous phase (only one Tg is measured for
24 3a <LOD <LOD
96 6 <LOD <LOD
the dispersions). Thermal analysis does not exclude
120 6 4a 3a the possibility that in addition to this amorphous
144 6 4a 3a phase, crystalline etravirine is present in the dis-
193 7 5a 5a persions. In the spray-dried powder, even at the
LOD, limit of detection. lowest assessed ratio (etravirine/HPMC 1:0.1), a
a
Estimated value (between the limit of detection and the limit of clear, single Tg is observed, suggesting that most
quantitation).
material is in the amorphous state. In order to assess
whether small amounts of crystalline drug substance
were present, other analytical techniques were

Figure 5. Overlay of reversing heat flow curves for etravirine/HPMC spray dried
formulations of different composition.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011
268 WEUTS ET AL.

patterns that are characteristic for crystalline etra-


virine (polymorph Form I). Even in these cases,
however, the XRD-patterns show only minor peaks
that are superimposed on a halo that originates from
the amorphous material present in the sample. These
XRD-analyses confirm the M-DSC-results, that is, the
1:0.3 and 1:0.1 formulations are predominantly
amorphous. FT-IR and FT-Raman analyses do not
indicate the presence of crystalline etravirine in any
of the dispersions. This suggests that these spectro-
scopic techniques are less sensitive than XRD in
detecting low levels of crystalline drug.
SS-NMR analysis was only performed on samples
containing an etravirine/HPMC ratio of 1:0.5, 1:1, and
1:3. The CP-MAS-13C-measurements supported the
conclusion that the drug was largely amorphous
in these samples.36,37 Specifically, proton T1 and T1r-
values were measured both for the drug and the
polymer (Tab. 5). The same was done for the physical
mixtures (PM) of amorphous (cryomilled) etravirine
and HPMC. In the solid dispersions, etravirine and
HPMC have similar T1 and T1r-values, pointing
to effective spin diffusion of the components and
miscibility. The physical mixtures on the other hand,
show a considerable difference in the 1H T1r-values of
Figure 6. Tg temperature-concentration profiles of
drug and polymer, confirming the heterogeneous
spray-dried powders of etravirine and HPMC (~ experi-
nature of these materials.
mentally determined values; & values calculated using the
GordonTaylor equation). Visualization techniques were also applied to better
characterize the spray-dried dispersions. Microscopic
applied including powder XRD, FT-IR/FT-Raman, images generated with FEG-ESEM (Fig. 8), indicate
and SS-NMR. that the dispersions consist of a mixture of spherical
Figure 3a and b shows an XRD overlay of the structures which were found to be hollow after
diffraction patterns of different dispersions. Only focused ion beam scanning electron microscopy
the spray-dried powders with a drug/polymer ratio (FIB-SEM) assessment. The size of these spheres
of 1:0.1 and 1:0.3 exhibit discernable diffraction and the extent to which they have collapsed is

Figure 7. Overlay of the total heat flow curves of etravirine/HPMC spray dried
powders at different compositions.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011 DOI 10.1002/jps
PREDICTION OF FORMULATION PROBABILITY OF SUCCESS FOR SOLID DISPERSIONS 269

Table 5. Relaxation Parameters of the Etravirine/HPMC in particular, is best predicated using a number of
Spray Dried Powders (SD) and Physical Mixtures (PM) early, efficient, and predictive screening experiments.
Measured with SS-NMR These include, but are not limited to, those which
1 1 probe physical stability. To configure a suitable
H T1 (s) H T1r (ms)
Sample roadmap, factors thought to contribute to, or detract
Etravirine/HPMC Etravirine HPMC Etravirine HPMC from stability need to be identified, assays need to be
developed and the aggregate information has to be
SD 1/0.5 1.43 1.47 14.72 14.59
SD 1/1 1.48 1.46 14.81 14.85 interpreted to provide an estimation of technical risk
SD 1/3 1.39 1.34 16.85 15.86 as well as advice on the best excipients and processing
PM 1/0.5 0.581 0.584 22.82 8.54 tools. The approach suggested herein includes three
PM 1/1 0.538 0.552 24.08 7.82 evaluation elements including: (1) an assessment of
PM 1/3 0.546 0.524 17.98 7.03
the pure amorphous phase, (2) an evaluation of
binary glasses cast from an appropriate solvent and
dependent on the etravirine/HPMC ratio and the (3) the study of processed powders using down-scaled
manufacturing process parameters. As mentioned production methods. The first component of this
in the Experimental Section, solvent composition, roadmap assesses the enablement likely required to
concentration of API and HPMC (and linked to this generate a pharmaceutically relevant dispersion. The
viscosity) were variables that were not the same for properties of the amorphous glass and its impact on
all prepared spray dried materials. It is well known the final dispersion depend on a number of factors
for instance that spray drying under wet conditions including the domain size in the dispersion and
(slow evaporation of the solvent) results in more miscibility, the possible molecular interaction of
collapsed spheres. Temperature settings were kept the amorphous phase with the glassy carrier and
constant for the preparation of the samples but the mixing Tg systems and storage conditions.911,29
the solvent composition was not. Having more or The greater the instability of the glass, the more
less of the most volatile solvent present in this important these stabilizing factors become. The
case would relate to more or less wet conditions. In data on etravirine presented here, as well as that
addition, the feed solutions that were used had presented in the literature16 indicate that it is difficult
different viscosities which impacts particle size. The to form a glass and once the glass is formed it is
more viscous the feed, the larger the particles that unstable even given its apparent high Tg and taking
are formed. No particles with other morphologies the Tg 50 rule into account.38,39 It is interesting to
were observed. compare these properties with those of another
In Figure 9, AFM images of etravirine powders, drug that has been marketed as a solid dispersion
ranging from high to low drug loading, are shown or solution. Itraconazole, for example, can be easily
both as 3D topography (odd numbered images) and converted to an amorphous phase using any number
phase-based renderings (even numbered images). of techniques including melt quenching, spray
The topography and phase images of the relatively drying, lyophilization rapid precipitation, milling,
low polymer-loaded spray-dried powders, that is, etc. (Tab. 2). Furthermore, even with a measured Tg
etravirine/HPMC 1:0.1 (i and ii) and 1:0.3 (iii and iv), of 598C, the prepared amorphous phase is stable to
show distinct phase-separated areas and evidence crystallization, even in open conditions for months to
of some ordered crystalline material. The AFM- years.4042 The stability of the amorphous form may
analyses confirm the predominantly amorphous be suggested by its Tg/Tm value of 0.76 (compared to
nature of these samples which was established with 0.69 for etravarine), its high energy of activation
the techniques outlined above (M-DSC, FT-Raman, for glassy to super-cooled liquid transition and by
FT-IR, and XRD). The AFM images also confirm the analysis using WilliamsWatts formalism where
presence of some crystalline drug as was suggested storage of the material at 308C below the Tg provide
by XRD. The spray-dried powder (drug/HPMC) for a t value of 245 days and when stored at 408C
1:0.5 (v and vi) show some phase-separated domains below Tg a t value of 80 years was calculated.41,42
but no crystalline material. Powders containing 1:1 to This stability is thought to be related to the ability of
1:9 (drug/HPMC) (images viixvi) all show homo- itraconazole to form a chiral nematic mesophase
genous topography with no indication of phase- (i.e., a two dimensionally structured glass) and this
separation or crystalline material. finding directly impacts dispersion robustness and
processing possibility.40,43,44 The practical conse-
quence of the relative stability of the itraconazole
DISCUSSION amorphous phase is that a number of formulation
concepts can increase oral bioavailability relative to
The optimal development strategy for solid dis- the crystalline drug including those based on com-
persions, in general, and the etravirine dispersion, plexation, bead coating, melt extrusion, spray-drying

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011
270 WEUTS ET AL.

Figure 8. FEG-ESEM images of etravirine/HPMC 1:1 (a and b), etravirine/HPMC 1:3


(c and d), etravirine/HPMC 1:6 (e and f) and broken particles to demonstrate the hollow
nature of the spheres (g and h).

and even tableting the amorphous form per se.4550 By therefore triggers a number of additional early
contrast, the properties of etravirine strongly suggest screens and assessments.
that the number of formulation options may be The examination of a cast film provides insight
limited and that stabilization of the amorphous phase into general miscibility and compatibility and pro-
will be paramount to achieving success. The data vides an easily testable, standardized matrix for
generated in this first part of the suggested roadmap assessing thermal properties, enthalpic relaxation

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011 DOI 10.1002/jps
PREDICTION OF FORMULATION PROBABILITY OF SUCCESS FOR SOLID DISPERSIONS 271

Figure 9. AFM-images of formulations etravirine/HPMC with ratios 1:0.1 (i and ii),


1:0.3 (iii and iv), 1:0.5 (v and vi), 1:1 (vii and viii), 1:2 (ix and x), 1:3 (xi and xii), 1:5 (xiii
and xiv), and 1:9 (xv and xvi).

(thermodynamic factors) and crystallization (kinetic A derivative parameter that can give insight into
factors)/phase separation within certain limits.31 dispersion stability is the extent of relaxation or the
Studies on etravirine and HPMC films pointed to degree to which the glass approaches its theoretical
the stabilizing effects of a HMPC-based dispersion on equilibrium condition (DH1 lim(t!1)DH which can
the drug glass and that increasing the polymer be estimated by DH1 (Tg  T)DCp).29,53,54 Relaxa-
component increased the relative stability. This was tion is assessed by annealing samples at different
manifested by an increasing Tg as the drug/polymer storage temperatures below the Tg and then asses-
ratio decreases, meaning that molecular mobility sing the change in relaxation enthalpy. Ordinarily
is reduced at a given storage temperature. The the enthalpy of relaxation increases with increasing
measured values (see the Results Section) deviate storage time and decreases with lower temperature in
from the theoretical Tgs calculated using the a nonlinear fashion. The WilliamsWatts relation-
GordonTaylor equation. The deviation could ship is a two parameter relationship (Ft 1  (DH/
have several causes including limited miscibility or DH1) exp  (t/t)b) that can suggest the time course
phase separation.51,52 Having considered this, the of the relaxation process allowing a prediction of
mixing Tgs, being intermediate between the Tgs of thermodynamic stability through the derived t (mean
HPMC and amorphous etravirine, suggested a drug/ relaxation time constant) value.11,55 Enthalpy of
polymer dispersion. Also, the fact that a discernable relaxation measurements for etravirine in cast
Tg for HPMC was not present suggests that a pure HPMC films suggested that there was a dramatic
polymer phase was absent. Another cause for this effect of polymer content and annealing temperature.
deviation may be the fact that the zero volume change In the 1:3 drug/polymer glasses, the extent of
on mixing assumption is not met.51,52 Violation from relaxation was 2%, 6%, and 22% after 24 days of
the free volume theory may be driven be differences storage at 45, 35, and 258C below Tg, respectively. For
in molecular interactions such that the homo- and the same conditions in the 1:1 drug/polymer system,
heteromolecular associations are different. these values were 30%, 55% and 61% demonstrating

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011
272 WEUTS ET AL.

the stabilizing effect of the polymer, the reduced same levels, often manifest different stability and
molecular mobility and expected long t value. biopharmaceutical properties based on how they were
However, it should be noted that thermodynamic generated.56,57 Examples of down-scaled processes
processes are only part of the picture and building a include the use of film forming or mini-fluid bed
dispersion only based on relaxation stability may be systems for coated bead systems, mini-extrusions for
inappropriate as the kinetic components may be more melt extrusion and the use of mini-spray-driers for
important depending on the particular system of this production technique. Similar to the approach
interest.32 Kinetically, factors that impede crystal- suggested in the case of the film study, data derived
lization include vitrification of the amorphous drug in from spray-dried powders indicated an increasing Tg
the glassy polymer preventing the formation of a with increasing polymer content which was asso-
crystalline lattice. In order to investigate this aspect ciated with higher stability and lower crystalline
of dispersion stability, the rate and extent of crystal- content (Figs. 5 and 6). As with the films, the data
lization were assessed. This was conducted at deviated from GordonTaylor behavior suggesting
temperatures at, or above the Tg, a domain where factors associated with homo- and heteromolecular
relaxation does not, theoretically, occur. These are forces may be in play. Interestingly, the measured
accelerated studies and the kinetic component of Tgs were comparable for the 1:1 spray-dried
crystallization from the dispersion may or may not drug/polymer mixture (1078C vs. 1068C for the cast
mimic that at ambient temperature, however these film) but tended to be progressively higher in value
approaches do provide a way to assess the effect of at the 1:2 (1138C (powder) vs. 1118C (film)) and
polymer content in the cast films on induction time 1:3 compositions (1188C (powder) vs. 1148C (film))
and crystallization extent. These parameters were lending some credence to the proposition that
estimated by preparing a standard curve for XRD the processing technique could play a role in
analysis. The data show that the 1:3 drug/polymer dispersion structure and performance. Factors that
systems crystallized more slowly than the 1:2 which may be germane in this regard may include the
in turn was more stable than the 1:1 drug/polymer efficiency of solvent evaporation or the nature of
dispersions. This is associated with the extent of drugpolymer mixing, that is, domain size and drug
crystallization at the end of the experiments (200 h) distribution.
for the 1:3 systems measured at Tg, Tg 10 and
Tg 25 of 5% (96 h indication time), 12% (8 h
indication time) and 31% (crystalline detected at CONCLUSIONS
the first sampling point), respectively. The compar-
able data for the 1:1 drug/polymer system was 7% (8 h Solid dispersions can allow for important drugs to
induction time), 25% and 43% (crystalline detected at reach the market by overcoming biopharmaceutical
the first sampling point in the latter two cases) at Tg, limitations imposed by NoyesWhitney and Ficks
Tg 10 and Tg 25, respectively. These experiments, First Law effects. Poor physical stability is one
based on the cast films, suggest that both thermo- factor that can limit the use of these potentially
dynamic (by a reduction in the enthalpy of relaxation enabling systems and whether this property impacts
with time) and kinetic (by reduced crystallization at dispersion applicability should be screened as early
and above the Tg) contributions could be responsible in the development cycle as possible to increase
for the stabilization of the formed dispersions.32 It is efficiency and probability of formulation success.
important to mention that while useful in screening Three inputs are suggested in making these afore-
mode, the cast films can contain large amounts of mentioned predictions including the stability and
residual solvent significantly affecting the mixing characteristics of the pure amorphous phase, the
Tgs and related properties. This suggests that these behavior of the amorphous drug when cast in films
data should be used qualitatively to suggest trends in containing a glassy carrier and when dispersed in a
drug stabilization. The advantage of this decision- polymer using a relevant process approach. The data
making element includes the fact that the approach suggest that etravirine is a poor glass former and the
is fast, automatable in a 96-well format and amorphous phase is unstable even given its high Tg.
information-rich, however they should not be over- By contrast, itraconazole manifests a much lower Tg
interpreted. and yet forms a particularly stable amorphous phase.
To better assess the role of processing and to While highly unstable as such, cast drug/HPMC films
provide a clearer view of the stability of the possible resulted in a dramatic stabilization of amorphous
formulation components, down-scaled techniques are etravirine with high polymer contents yielding
suggested to prepare powders for analytical scrutiny. progressively more stable dispersions. This informa-
This last component of the evaluation roadmap is tion may point to the fact that multiple dispersion
important given that many amorphous dispersions, formulation are possible with itraconazole while only
even when containing the same excipients at the the spray-dried dosage form provided good oral

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011 DOI 10.1002/jps
PREDICTION OF FORMULATION PROBABILITY OF SUCCESS FOR SOLID DISPERSIONS 273

bioavailability for etravirine (when compared to other separation in solid dispersions of Itraconazole and Eudragit1
amorphous systems). The generation and study of E100 using microthermal analysis. Pharm Res 20:135138.
19. Bauer-Brandl A. 1996. Polymorphic transition of cimetidine
spray-dried powders allowed for a useful under-
during manufacture of solid dosage forms. Int J Pharm 140:
standing of the formulation elements such that the 195206.
ultimate formulation could be optimally designed and 20. Shi HG, Farber L, Michaels JN, Dickey A, Thompson KC,
produced. Shekular SD, Hurter PN, Reynolds SD, Kaufman MJ. 2003.
Characterization of crystalline drug nanoparticles using
atomic force microscopy and complementary techniques.
REFERENCES Pharm Res 20:479484.
21. Zhang J, Ebbens S, Chen X, Jin Z, Luk S, Madden C, Patel N,
1. Lipinski CA. 2000. Drug-like properties and the causes of poor Roberts CJ. 2006. Determination of the surface free energy of
solubility and poor permeability. J Pharmacol Toxicol Methods crystalline and amorphous lactose by atomic force microscopy
44:235249. adhesion measurements. Pharm Res 23:401407.
2. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. 2001. 22. Ward S, Perkins M, Zhang J, Roberts CJ, Madden CE, Luk SY,
Experimental and computatorial approaches to estimate solu- Patel N, Ebbens SJ. 2005. Identifying and mapping surface
bility and permeability in drug discovery and development amorphous domains. Pharm Res 22:11951202.
settings. Adv Drug Deliv Rev 46:326. 23. Price R, Young PM. 2004. Visualization of the crystallisation of
3. Lipinski CA. 2001. Avoiding investment in doomed drugs. Curr lactose from the amorphous state. J Pharm Sci 93:155164.
Drug Discov 1:1719. 24. Hancock BC, Zografi G. 1997. Characteristics and significance
4. Lipinski C, Hopkins A. 2004. Navigating chemical space for of the amorphous state in pharmaceutical systems. J Pharm Sci
biology and medicine. Nature 432:855862. 86:112.
5. Lipinski CA. 2006. The anti-intellectual effect of intellectual 25. Yu L. 2001. Amorphous pharmaceutical solids: Preparation,
property. Curr Opin Chem Biol 10:380383. characterization and stabilization. Adv Drug Deliv Rev 48:
6. Liu R. 2000. Water insoluble drug formulation. Interpharm 2742.
Press. Denver, CO. 26. Ping G. 2008. Amorphous pharmaceutical solids: Character-
7. Dressman J, Butler J, Hempenstall J, Reppas C. 2001. The ization, stabilization and development of marketable formula-
BCS: Where do we go from here? Pharm Tech 25:6876. tions of poorly water soluble drugs with improved oral
8. Brewster M, Loftsson T. 2007. Cyclodextrins as pharmaceutical absorption. Mol Pharm 5:903904.
solubilizers. Adv Drug Deliv Rev 59:6456766. 27. Mathot VBF. 2001. New routes for thermal analysis and calori-
9. Leuner C, Dressman J. 2000. Improving drug solubility for oral metry as applied to polymeric systems. J Therm Anal Cal 64:
delivery using solid dispersion. Eur J Pharm Biopharm 50: 1535.
4760. 28. Chawla G, Bansal AK. 2007. A comparative assessment of
10. Serajuddin AT. 1999. Solid dispersion of poorly water-soluble solubility advantage from glassy and crystalline forms of a
drugs. Early promises, subsequent problems and recent break- water-insoluble drug. Eur J Pharm Sci 32:4557.
throughs. J Pharm Sci 88:10581066. 29. Crowley KJ, Zografi G. 2001. The use of thermal methods for
11. Hancock BC. 2002. Disordered drug delivery: Destiny, predicting glass-former fragility. Thermochim Acta 380:79
dynamics and the Deborah number. J Pharm Pharmacol 54: 93.
737746. 30. Wang L, Angell CA, Richert R. 2006. Fragility and thermo-
12. Andries K, Azijn H, Thielemans T, Ludovici D, Kukla M, dynamics in nonpolymeric glass-forming liquids. J Chem Phys
Heeres J, Janssen P, De Corte B, Vingerhoets J, Pauwels R, 125:074505.
de Bethune MP. 2004. TMC125, a novel next-generation non- 31. Verreck G, Six K, Van den Mooter G, Baert L, Peeters J,
nucleoside reverse transcriptase inhibitor active against non- Brewster ME. 2003. Characterization of solid dispersions of
nucleoside reverse tgranscriptase inhibitor-resistant human itraconazole and hydroxypropylmethylcellulose prepared by
immunodeficiency virus type 1. Antimicrob Agents Chemother melt extrusionPart 1. Int J Pharm 251:165174.
48:46804686. 32. Bhugra C, Pikal MJ. 2008. Role of thermodynamic, molecular
13. Vingerhoets J, Azijn H, Fransen E, De Baere I, Smeulders L, and kinetic factors in crystallization from the amorphous state.
Jochmans D, Andries K, Pauwels R, de Bethune MP. 2005. J Pharm Sci 97:13291349.
TMC125 displays a high genetic barrier to the development 33. Gordon M, Taylor JS. 1952. Ideal copolymers and the second-
of resistance: Evidence from in vitro selection experiments. order transitions of synthetic rubbers. I. Non-crystalline copo-
J Virol 79:1277312782. lymers. J Appl Chem 2:493501.
14. Habib MJ. 2000. Fundamentals of solid dispersions. In: Habib 34. Simha R, Boyer RF. 1962. On a general relation involving the
MJ, editor. Pharmaceutical solid dispersion technology. Boca glass temperature and coefficients of expansion of polymers.
Raton, FL: CRC Press. pp 736. J Chem Phys 37:10031007.
15. Craig DQM. 2002. The mechanisms of drug release from solid 35. Van den Mooter G, Van den Brande J, Augustijns P, Kinget R.
dispersions in water-soluble polymers. Int J Pharm 231:131 1999. Glass forming properties of benzodiazepines and co-eva-
144. porate systems with poly(hydroxyethyl methacrylate). J Therm
16. Qi S, Weuts I, De Cort S, Stokbroekx S, Leemans R, Reading M, Anal Calor 57:493507.
Belton P, Craig DQM. 2009. An investigation into the crystal- 36. Lubach J, Xu D, Segmuller B, Munson EJ. 2007. Investigation
lization behavior of an amorphous cryomilled pharmaceutical of the effects of pharmaceutical processing upon solid-state
material above and below the glass transition. J Pharm Sci NMR relaxation times and implications to solid-state stability.
99:196208. J Pharm Sci 96:777787.
17. Shah B, Kakumanu VK, Bansal AK. 2006. Analytical techni- 37. Geppi M, Mollica G, Borsacchi S, Veracini CA. 2008. Solid-state
ques for quantification of amorphous/crystalline phases in NMR studies for pharmaceutical systems. Appl Spectrosc Rev
pharmaceutical solids. J Pharm Sci 95:16411665. 43:202302.
18. Six K, Murphy J, Weuts I, Craig DQM, Verreck G, Peeters J, 38. Hancock BC, Shamlin SL, Zografi G. 1995. Molecular mobility
Brewster M, Van den Mooter G. 2003. Identification of phase of glassy pharmaceutical solids. Pharm Res 12:799806.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011
274 WEUTS ET AL.

39. Franks F. 2003. Scientific and technological aspects of aqueous 49. Cha B, Oh J. 1998. An itraconazole exhibition an improved
glasses. Biophys Chem 105:251261. solubility, a method of preparing the same a pharmaceutical
40. Six K, Verreck G, Peeters J, Binnemans K, Berghmans H, composition for oral administration comprising the same.
Augustijns P, Kinget R, Van den Mooter G. 2001. Investigation WO9857967.
of thermal properties of glassy itraconazole: Identification of a 50. Peeters J, Verreck G, Vandecruys R, Brewster M. 2008.
monotropic mesophase. Thermochim Acta 376:175181. Dissolution and human bioavailability in supersaturating drug
41. Six K, Verreck G, Peeters J, Augustijns P, Kinget R, Van den delivery systems: Formulations of itraconazole. 2008 American
Mooter G. 2001. Characterization of glassy itraconazole: A Association of Pharmaceutical Scientists (AAPS) Annual Meet-
comparative study of its molecular mobility below Tg with that ing and Exposition, Abstract No. T2097.
of structural analogs with MTDSC. Int J Pharm 213:163173. 51. Tajber L, Corrigan OI, Healy AM. 2005. Physicochemical eva-
42. Weuts I, Kempen D, Six K, Peeters J, Verreck G, Brewster M, luation of PVP-thiazide diuretic interactions in co-spray dried
Van den Mooter G. 2003. Evaluation of different calorimetric compositesAnalysis of glass transition composition relation-
methods to determine the glass transition temperature and ships. Eur J Pharm Sci 24:553563.
molecular mobility below Tg for amorphous drugs. Int J Pharm 52. Gupta P, Bansal AK. 2005. Molecular interactions in celecoxib-
259:1725. PVP-meglumine amorphous systems. J Pharm Pharmacol 57:
43. Bunjes H, Rades T. 2005. Thermotropic liquid crystalline 303310.
drugs. J Pharm Pharmacol 57:807816. 53. Craig DQM, Royall PG, Kett VL, Hopton ML. 1999. The rele-
44. Stevenson CL, Bennett DB, Lechuga-Ballesteros D. 2005. vance of the amorphous state to pharmaceutical dosage forms:
Pharmaceutical liquid crystals: The relevance of partially Glassy drugs and freeze dried systems. Int J Pharm 179:179
ordered systems. J Pharm Sci 94:18611880. 207.
45. Gilis PA, De Conde V, Vandecruys R. 1997. Beads having a core 54. Kawakami K, Pikal MJ. 2005. Calorimetric investigation
coated with an antifungal and a polymer. US patent 5633015. of the structural relaxation of amorphous materials: Evaluat-
46. Brewster ME, Vandecruys R, Verreck G, Noppe M, Peeters J. ing validity of the methodologies. J Pharm Sci 94:948
2002. A novel cyclodextrin-containing glass thermoplastic sys- 965.
tems (GTS) for formulating poorly water soluble drug candi- 55. Shamblin SL, Hancock BC, Dupuis Y, Pikal MJ. 2000. Inter-
dates: Preclinical and clinical results. J Incl Phenom Macro pretation of relaxation time constants for amorphous pharma-
Chem 44:3538. ceutical systems. J Pharm Sci 89:417427.
47. Six K, Berghmans H, Leuner C, Dressman J, Van Werde K, 56. Weuts I, Kempen D, Decorte A, Verreck G, Peeters J, Brewster
Mullens J, Benoist L, Thimon M, Meublat L, Verreck G, Peeters M, Van den Mooter G. 2004. Phase behavior analysis of solid
J, Brewster M, Van den Mooter G. 2003. Characterization of dispersions of loperamide and two structurally related
solid dispersions of itraconazole and hydroxypropylmethyl-cel- compounds with the polymers PVP-K30 and PVP-VA64. Eur
lulose prepared by melt extrusionPart II. Pharm Res 20: J Pharm Sci 22:375385.
10471054. 57. Weuts I, Kempen D, Decorte A, Verreck G, Peeters J, Brewster
48. Peeters J, Neeskens P, Tollenaere JP, Van Remoortere P, M, Van den Mooter G. 2005. Physical stability of the amorphous
Brewster ME. 2002. Characterization of the interaction of state of loperamide and two fragment molecules in solid
2-hydroxypropyl-b-cyclodextrin with itraconazole at pH 2, dispersions with the polymers PVP-K30 and PVA-VA64. Eur
4 and 7. J Pharm Sci 91:14141422. J Pharm Sci 25:313320.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 1, JANUARY 2011 DOI 10.1002/jps

Anda mungkin juga menyukai