SlideShare a Scribd company logo
1 of 10
Download to read offline
Full Terms & Conditions of access and use can be found at
http://www.tandfonline.com/action/journalInformation?journalCode=iddi20
Download by: [LIU Libraries] Date: 05 May 2016, At: 07:09
Drug Development and Industrial Pharmacy
ISSN: 0363-9045 (Print) 1520-5762 (Online) Journal homepage: http://www.tandfonline.com/loi/iddi20
Influence of spray drying and dispersing agent on
surface and dissolution properties of griseofulvin
micro and nanocrystals
Dhaval A. Shah, Manan Patel, Sharad B. Murdande & Rutesh H. Dave
To cite this article: Dhaval A. Shah, Manan Patel, Sharad B. Murdande & Rutesh H. Dave
(2016): Influence of spray drying and dispersing agent on surface and dissolution properties
of griseofulvin micro and nanocrystals, Drug Development and Industrial Pharmacy, DOI:
10.1080/03639045.2016.1178770
To link to this article: http://dx.doi.org/10.1080/03639045.2016.1178770
Accepted author version posted online: 14
Apr 2016.
Published online: 04 May 2016.
Submit your article to this journal
Article views: 13
View related articles
View Crossmark data
RESEARCH ARTICLE
Influence of spray drying and dispersing agent on surface and dissolution
properties of griseofulvin micro and nanocrystals
Dhaval A. Shaha
, Manan Patela
, Sharad B. Murdandeb
and Rutesh H. Davea
a
Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA; b
Drug Product Design, Pfizer
Worldwide R&D, Groton, CT, USA
ABSTRACT
The purpose for the current research is to compare and evaluate physiochemical properties of spray-dried
(SD) microcrystals (MCs), nanocrystals (NCs), and nanocrystals with a dispersion agent (NCm) from a poorly
soluble compound. The characterization was carried out by performing size and surface analysis, interfacial
tension (at particle moisture interface), and in-vitro drug dissolution rate experiments. Nanosuspensions
were prepared by media milling and were spray-dried. The SD powders that were obtained were character-
ized morphologically using scanning electron microscopy (SEM), polarized light microscopy (PLM), and
Flowchem. Solid-state characterization was performed using X-ray powder diffraction (XRPD), Fourier trans-
fer infrared spectroscopy (FT-IR), and differential scanning calorimetry (DSC) for the identification of the
crystalline nature of all the SD powders. The powders were characterized for their redispersion tendency in
the water and in pH 1.2. Significant differences in redispersion were noted for both the NCs in both dissol-
ution media. The interfacial tension for particle moisture interface was determined by applying the BET
(Braunauer–Emmett–Teller) equation to the vapor sorption data. No significant reduction in the interfacial
tension was observed between MCs and NCs; however, a significant reduction in the interfacial tension was
observed for NCm at both 25 
C and 35 
C temperatures. The difference in interfacial tension and redisper-
sion behavior can be attributed to a difference in the wetting tendency for all the SD powders. The dissol-
ution studies were carried out under sink and under non-sink conditions. The non-sink dissolution
approach was found suitable for quantification of the dissolution rate enhancement, and also for providing
the rank order to the SD formulations.
ARTICLE HISTORY
Received 29 December 2015
Revised 30 March 2016
Accepted 7 April 2016
Published online 3 May 2016
KEYWORDS
Griseofulvin; interfacial
tension; nanocrystals; non-
sink dissolution;
redispersion; spray dryer
Introduction
It has been reported that more than half of the identified potential
drug candidates have challenges related to aqueous solubility and/
or the dissolution rate1
. A majority of these compounds belong to
the BCS class II and IV categories2,3
. The rate and the extent of
absorption of the Class II compounds are highly dependent on the
performance of the formulated product4–6
. Several formulation
approaches have been suggested and reported to overcome these
problems. The formulation of nanocrystals (NCs; suspension or
solid) is one of the preferred approaches that has been used for
the drugs which belong to the above BCS classes7,8
. NCs offer sev-
eral advantages, including enhancement in bioavailability, reduc-
tion in therapeutic dose, and elimination of the food effect9–14
.
The solubility behavior of crystalline nanoparticles can be
explained by the Ostwald–Frendulich equation. Based on the pub-
lished reports, the thermodynamic solubility enhancement of nano-
crystalline particles should be approximately 10–15%8,15,16
. On the
other hand, nanosuspensions have a very high surface-to-volume
ratio and high Gibbs energy, which indicates the instability of the
system. They tend to form aggregates due to Ostwald’s ripen-
ing13,17
. Nanosuspensions can be stabilized and their shelf life can
be enhanced by converting them into solid powders by spray dry-
ing and/or freeze drying10,18
.
Upon solidification, the surface properties of dried particles (i.e.
size, shape) may tend to change significantly, which will have a dir-
ect impact on the wetting and dissolution performance of the
formulation19
. Hence, it is crucial to assess the surface and the
interfacial properties of the dried particles20,21
. It is also known that
these suspensions (micro/nano) may form reversible or nonreversi-
ble aggregates upon solidification. Therefore, aggregation is
another key product quality parameter that needs to be assessed
for effectiveness with the dried particles22,23
. During spray drying,
the particle size distribution is governed by the nozzle size24
. It is
possible to have different dissolution behavior for formulations
with the same particle size due to change in surface wetting prop-
erty25
. Therefore, it will be important to determine the role of the
interfacial tension (at particle to moisture interface) in governing
the redispersion and the kinetic solubility (initial dissolution rate)
of NCs and micro crystals (MCs).
In the current study, we have formulated spray-dried (SD) nano-
crystalline powder (with and without the bulking agent mannitol)
and MCs of a poorly soluble drug. Griseofulvin (a BCS Class II com-
pound) was selected as a model compound and docusate sodium
(ionic stabilizer) and HPC-SL (Hydroxypropyl cellulose-low viscosity
grade; polymeric stabilizer) were utilized to prevent agglomeration
of nanosuspensions before spray drying, while the mannitol was
used as a dispersing agent for spray drying. The SD powders were
evaluated for surface characterization (size and shape analysis),
solid-state characterization (XRPD, DSC, and FT-IR), interfacial ten-
sion (DVSO), redispersion, and in-vitro dissolution rates (sink and
non-sink). This study shows the effect of interfacial tension in gov-
erning the dissolution of NCs and quantitates the dissolution rate
CONTACT Rutesh H. Dave rutesh.dave@liu.edu Arnold  Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201,
USA.
ß 2016 Informa UK Limited, trading as Taylor  Francis Group
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY, 2016
http://dx.doi.org/10.1080/03639045.2016.1178770
Downloadedby[LIULibraries]at07:0905May2016
enhancement for the SD powders using non-sink conditions to
provide them in rank order based on their in-vitro performance.
Materials
Griseofulvin was obtained from Spectrum Chemicals (New
Brunswick, NJ). The yttrium-stabilized zirconium oxide beads were
obtained from Glen Mills Inc. (Clifton, NJ). Hydroxypropylcellulose
(HPC)–SL was obtained from Nisso America Inc. (New York, NY).
Other laboratory chemicals, such as mannitol, docusate sodium
USP, acetonitrile, and trifluoroacetic acid (TFA), were purchased
from Spectrum Chemicals (New Brunswick, NJ) and were of analyt-
ical grades. All the excipients utilized in this study were of high
purity. The chemical structure of API and excipients are presented
in Figure 1.
Methods
Wet milling
Griseofulvin (50 mg/mL) was suspended in an aqueous stabilizer
solution which is a mixture of 1.25% (w/v) HPC–SL (stearic stabil-
izer) and 0.05% (w/v) docusate sodium (ionic stabilizer). The sus-
pension (500 mL) was milled using the yttrium stabilized zirconium
oxide beads (YTZ beads) at a milling speed of 1000 rpm in the con-
tinuous model8
. The suspension formulation was milled for 4 h,
and the particle size was measured during and following the mill-
ing process. The temperature of the sample was maintained below
25 
C using the cold water that surrounded the milling chamber to
compensate for the heat generated by high-speed milling. The sus-
pension was harvested by filtration through a coarse sintered-glass
funnel filter (75 mm). The nanosuspension was divided into two
equal parts, and in one part the mannitol was dissolved in a 1:2
ratio (Griseofulvin: mannitol). The suspension was then stirred for
at least 30 min and then prepared for spray drying. The microcrys-
talline suspension was prepared using the same method and the
same stabilizer concentration with the absence of the milling
vehicle (i.e. YTZ beads).
Spray drying
Nano (with and without the mannitol) and microcrystalline suspen-
sions were spray-dried using a B-290 spray dryer (Buchi
Labortechhnik AG, Flawil, Switzerland). The spray dryer was equili-
brated using distilled water at 128 
C inlet temperature, 3 mL/min
feed rate, and 100% aspiration rate. The outlet temperature was
approximately 75 
C. The aforementioned SD parameters were
determined from some preliminary experiments. Once the spray
dryer was equilibrated, the suspension formulations were subjected
to spray drying with continuous stirring using a magnetic stirrer.
SD powders were collected from the collection chamber and
immediately analyzed for particle size, process yield, and
crystallinity.
Particle size, surface and shape analysis
A dynamic light scattering (DLS) instrument (Malvern Instruments
Inc., Malvern, UK) was used to measure particle size and the size
distribution of suspensions. All three SD powders were subjected
to particle size and shape analysis. Particle size measurements of
SD powders were performed using a Sympatec GmbH System-
Partikel-Technik (Sympatec Inc., Pennington, NJ). Briefly, around
100 mg of a sample was subjected for analysis. Compressed air
was used to disperse the powder, which is blown through the laser
beam and collected by suction after the analysis. Polarized light
microscopy (PLM; Model BX60FS; Olympus BX60, Center Valley, PA)
and scanning electron microscopy (SEM, ASPEX Scanning Electron
Microscope model PSEM 3025 SEM/EDS, Fei Inc., Hillsboro, OR)
were conducted to observe the surface morphology and texture of
the pure materials and SD powders. The polarized microscopic
images were taken using a camera from Diagnostic Instruments
Inc. (Sterling Heights, MI), and the analysis was performed with the
help of SPOTTM
imaging advanced software. For the SEM imaging,
the samples were mounted on double-sided carbon tape. The low
vacuum (LV) mode was used to prevent the samples from charg-
ing. The energy beam and filament drive were adjusted to 20 kev
and 72%, respectively. The samples were analyzed under approxi-
mately 60% contrast and 30% brightness in secondary electron
mode. Particle shape and further morphological characterization
were performed using the flow cam machine (Fluid Imaging
Technologies, Scarborough, ME). Here, the sample was prepared by
dispersing the SD powders into the water and then subjected to
analysis. The machine captures particle images at up to 22 frames
per second. The analysis was performed by keeping the flow rate
at 0.3 mL/min and viewed under 4Â optical lens. The morpho-
logical measurements (i.e. shape) of particles were calculated using
the Visual spread sheetVR
Particle analysis software.
Differential scanning calorimetry
Modulated differential scanning calorimetric (mDSC) analysis was
performed using a Q200 (TA Instruments, New Castle, DE)
equipped with a cooling system. A sample (5–10 mg) was weighed
into an aluminum pan with a pin hole and hermitically sealed. The
DSC parameters used were: a sample equilibration at 30 
C, modu-
lated for ±0.318 
C every 60 s, isothermal for 5 min and heated to
230 
C with a ramp of 2 
C/min. The DSC cell was purged with
nitrogen gas at a flow rate of 50 mL/min. The data obtained were
analyzed with the Universal Analysis Software (TA Instruments,
New Castle, DE).
Powder X-ray diffraction
Powder X-ray diffraction was done using a scanning diffractometer
(Advanced Diffraction System, Scintag Inc. Model XI, Cupertino,
CA), controlled by a computer with diffraction management system
software for Windows NT. The radiation used was generated by a
copper Ka filter, with a wavelength of 1.54 A
at 45 kV and 40 mA.
High energy mixture patterns obtained by the X-ray were
Figure 1. Chemical structures of a model compound and excipients.
2 D. A. SHAH ET AL.
Downloadedby[LIULibraries]at07:0905May2016
compared to a reference drug to confirm crystallinity26
. Samples
were scanned over a range of 5–40
degrees, using a scan rate of
1
/min and a step size of 0.02. X-ray diffraction patterns were
obtained for pure griseofulvin, excipients, physical mixtures, and
formulated SD powders. Data analysis for recorded patterns was
done using X’Pert Data Viewer software.
Infrared spectroscopy (FT-IR)
FTIR spectra for all the abovementioned samples were obtained by
Nicolet-FTIR equipped with smart ATR (Thermo Fisher Scientific Inc.
Madison, WI), and a DTGS-KBr (Deuterated triglycine sulfate)
detector was used for background and baseline corrections. Small
amounts of the sample were placed on the diamond crystal of the
ATR and then the ATR knob was closed. Finally, the spectra were
collected from 400 to 4000 cmÀ1
with 264 accumulations at a reso-
lution of 4 cmÀ1
.
Gravimetric vapor sorption and interfacial energy (DVS)
Moisture sorption profiles were obtained for the SD powders using
a SGA-100 Sorption Analyzer (VTI Corporation, Hialeah, FL). The
measurements were taken at 25 
C (storage temperature) and
35 
C (close to physiological temperature) using a sample size of
10–20 mg. All SD samples were exposed to an automated compu-
terized sequence of increasing relative humidity (RH) in steps of
10% RH. The RH range was from 0% RH to 95% RH. The vapor
sorption analysis was carried out to calculate the interfacial tension
and water uptake capacity of all three powders as described in the
literature27
. The adsorption of moisture onto multilayer surfaces
is believed to follow the Brunauer–Emmett–Teller (BET) model.
Similarly, we made the assumption that adsorption of moisture
onto SD powder particles would follow the BET multilayer model.
The BET constants (m0 and C) were determined from the initial lin-
ear region of the moisture sorption adsorption isotherm (0.35%
w/w). The interfacial tension at particle moisture interface was
determined by calculating the binding energy (from BET constant
C) and the average surface area of a water molecule (Appendix A).
HPLC analytical method
The quantitative analysis of model compounds used in the solubil-
ity studies was performed by a reverse-phase gradient HPLC using
an Agilent 1100 System (Agilent Technologies, Palo Alto, CA)
equipped with a 4.6 Â 75 mm, 2.7 mm C18 column (Advanced
Materials Technology, Wilmington, DE). The temperature of the col-
umn was kept at 45 
C, and the wavelength of detection was set
to 210 nm. The mobile phase consisted of a binary gradient of
Solvent A (acetonitrile) and Solvent B (HPLC grade water with
0.05% TFA). The linear gradient started at 5% A and increased to
95% A in 8.5 min, followed by a return to the starting condition
within 1.6 min and equilibrated at the starting condition for 2 min.
The injection volume was 10 mL. The flow rate was 1.0 mL/min, and
the total run time was set for 13.10 min. This reverse-phase gradi-
ent method provided a baseline resolution and excellent peak
characteristics for the griseofulvin. Quantification of drug concen-
trations was performed by analyzing the peak area at retention
using Agilent ChemstationVR
analytical software.
Redispersion analysis
The nanosuspensions have tendency to form loose or hard aggre-
gates when spray-dried. Sometimes these aggregates are reversible
and sometime they are not reversible. To evaluate this behavior,
we carried out the redispersion study of SD NCs (with and without
mannitol) in the DI water and in the simulated gastric media (pH
1.2). The study was performed in a conventional USP-type – II dis-
solution apparatus at 100 rpm and 37 
C temperature. Here, sam-
ples were withdrawn at different time intervals and subjected to
the particle size analysis using the zetasizer28
.
In vitro dissolution rate determination
A dissolution apparatus (Distek 2100C; Distek Inc., North Brunswick,
NJ) equipped with a USP-type-II (paddle) was used to measure the
dissolution rates of crystalline SD dispersions. The dissolution study
was carried out by comparing the initial dissolution rates of MCs
and NCs (with and without mannitol). Each dissolution study was
carried out at 37 ± 0.5 
C under a 100-rpm paddle speed in a USP-
type II apparatus for 2 h. The dissolution was carried out under
sink condition (sink condition factor 5.7Â), and under non-sink
conditions (at 20%, 50%, and 90% saturation level of API in dissol-
ution media) with a 50 mg loading of griseofulvin. A 10 mL of a dis-
solution sample was withdrawn at intervals of 0, 2, 5, 7, 10, 20, 30,
45, 60, 90, and 120 min and replaced with the same volume of dis-
solution media (pre-equilibrated at 37 ± 0.5
). Samples were filtered
through 0.1-mm (MCs) and 0.02-mm (NCs) anotop syringe filters
(WhatmanTM
; GE Healthcare, Piscataway, NJ) and subjected to
HPLC analysis. The first few milliliters of each sample were dis-
carded to minimize losses because of adsorption. The discarded
volume selected for each compound was determined by a separate
filter validation experiment (data not shown). All studies were per-
formed in triplicate29–35
.
Results and discussion
Morphological characterization
The particle size of the suspensions before the spray dying was
measured using DLS. The particle sizes of the SD powders were
measured using the Sympatec GmbH System-Partikel-Technik
(HELOS  RODOS). The particle size and shape analysis data are
presented in Table 1. The particle size of micronized SD powder
upon drying is approximately 14 mm, while the particle size of
both SD nanosuspension powders upon drying are around 6 mm.
The particle size of nano powders are significantly lower than the
particle size of the micronized powder, which is explainable due
to difference in particle size before spray drying. There is no sig-
nificant difference in particle size between nanosuspensions
with and without mannitol. Morphological characterization by
PLM and SEM (Figures 2 and 3) shows the particle size difference
between MCs and NCs. Moreover, the NCs in both formulations
became hollow upon particle size reduction while MCs appear
cylindrical shaped. The SEM image of nanocrystals with mannitol
(NCm) also shows the presence of mannitol on the surface of
NCs. Initial observation from SEM images of the NCs with and
without the mannitol showed formation of aggregates upon
spray drying. Therefore, redispersion analysis was carried out for
further characterization of the aggregation property (reversible or
irreversible). The FLOWCHEM machine has the ability to capture
30 unique particle measurements. The morphological measure-
ments for SD particles were calculated using Visual spread sheetVR
particle analysis software. The NCm were found to be spherical in
shape as compared to NCs without the mannitol and the MCs
(Table 1).
Solid-state characterization
It is known that changes in the crystalline state can occur during
both milling and spray drying. To ensure that the changes are due
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 3
Downloadedby[LIULibraries]at07:0905May2016
to particle size reduction and that no amorphous griseofulvin was
formed, MDSC, XRPD, and FT-IR were performed on the SD
samples.
Griseofulvin showed distinct crystalline peaks for 2h at 11
and
17
(Figure 4(a)) in all the SD powders. As shown by the XRPD pat-
terns (Figure 4(a)), the reduction in the intensity and broadening of
Figure 2. PLM images of (a) pure griseofulvin, (b) MC, (c) NCs, and (d) NCm.
Figure 3. SEM images of (a) HPC-SL, (b) DOSS, (c) mannitol, (d) pure griseofulvin, (e) MC, (f) NCs, and (g) NCm.
Table 1. Particle size and shape analysis.
Formulation
Avg. particle size
before spray drying PDI
Avg. particle size
after SD (lm) Span(D90–D10)/D50
Circularity
(Hu)
Aggregation based
on SEM/PLM
MC 11 lm – 14.9 2.3 0.81 Yes
NCs 210 nm 0.210 6.64 1.85 0.82 Yes
Nanocrystal-mannitol 205 nm 0.192 6.42 1.82 0.95 Yes
4 D. A. SHAH ET AL.
Downloadedby[LIULibraries]at07:0905May2016
the peaks is observed for the nanoparticles. XPRD of SD powders
showed a small hump in between 20 and 30 at 2h scale represents
possible minor amorphous conversion. Figure 4(b) shows the MDSC
thermograms for pure griseofulvin and the SD formulations. The
thermograms were compared to confirm the crystallinity upon mill-
ing and spray drying. The onset melting temperature of griseofulvin
and mannitol were detected at 219.0 
C and 168.4 
C, respectively.
After milling and spray drying, a reduction in the onset of the melt-
ing temperature (2–4 
C) and the heat of fusion was observed for
both griseofulvin and mannitol. The melting peaks of griseofulvin
shifted in the SD formulations (Figure 4(b)). The observed melting
point reduction and peak shift have been reported to be in agree-
ment with the particle size reduction. It can also be explained by
the Gibbs–Thomson equation36
, which explains that, the melting
temperature of a material is proportional to its cohesive energy.
Since the atoms at the surface have reduced cohesive energy com-
pared to bulk material due to physical milling and spray drying.
Therefore, they require less energy to free from the solid phase (i.e.
melting), which results in the reduction of melting point. Based on
the MDSC and XRPD data, it can be concluded that the wet-media
milling and spray drying might have affected the crystalline struc-
ture of the model compound by partially (minor) converting it in to
the amorphous form. For further clarification of the crystalline struc-
ture, FT-IR spectroscopy was conducted on the SD samples (Data
not shown). The SD formulation with mannitol showed intense peak
at 3288À1
cm and 3388À1
cm (–OH stretching) suggesting the pres-
ence of mannitol in the formulation. These transmissions were
absent in the IR spectra of MCs and NCs without the mannitol. The
IR spectra of all three SD formulations are same except the increase
in the –OH and –CH stretching intensity due to the presence of
mannitol in the SD formulation with mannitol.
Interfacial energy and water uptake tendency
The dynamic vapor sorption analysis was performed to evaluate the
moisture uptake tendency of the SD powders. The water uptake
tendency of NCm was found to be higher compared to both the
MCs and NCs. For a detailed evaluation of this, we calculated the
interfacial energy as described in Appendix A, and the results are
presented in Table 2. For the colloidal dispersion systems, the inter-
facial energy plays an important role. For different compounds, the
solubility and, eventually, the dissolution rate increased as the sol-
id–liquid interfacial tension decreases37
. The interfacial tension val-
ues for the MC and NC do not have any significant difference at
either temperature (both at 25 
C and 35 
C). The NCm showed less
interfacial tension values compared to MCs and NCs without manni-
tol at both temperatures. Here, mannitol has a characteristic prop-
erty to act as a solubilizing and dispersing agent. Mannitol acts by
forming capillaries in contact with aqueous solutions and generates
rapid dispersion and dissolution38
. Based on these studies, we can
conclude that the dissolution and redispersion behavior for NCm
will be significantly higher due to the enhancement of the wetting
property and a reduction in interfacial tension. Crisp39
and his col-
leagues have also reported that the dissolution of NCs is more
dependent on interfacial tension than the surface area.
Redispersion analysis
The nanosuspensions have a tendency to form loose or hard
aggregates when SD. Nanoparticle aggregation was visually
observed for SD powders in SEM images. Sometimes these aggre-
gates are reversible and sometime they are not reversible. In order
to assess their aggregation property, the redispersion analysis was
carried out. The SD powder of NCm redispersed quickly during the
redispersion study in water and in pH 1.2 (Figure 5(a) and (b)). The
NCm quickly reached the initial particle size. On the other hand,
the NCs without the mannitol were not able to achieve the initial
particle size (200–300 nm) when spray-dried. The dispersion behav-
ior of particles in the liquid phase is mainly affected by the inter-
action between the particles and the dispersion media. Therefore,
the presence of mannitol aids in wetting the particles quickly by
forming reversible aggregates. In the case of the NCs without the
Figure 4. Solid-state characterization and comparison of pure API, physical mixture, MCs, NCs, and NCm (a) XRPD, and (b) DSC.
Table 2. Determination of BET constants and interfacial tension.
Formulation Particle size (lm)
C
(binding constant)
m0
(monolayer g/g
solids)
DE (J/mole) (sur-
face binding
energy)
c
(mN/m) (interfacial
tension)
25 
C 35 
C 25 
C 35 
C 25 
C 35 
C 25 
C 35 
C
MC 14.9 À20.58 À20.43 12.15 1.77 7492 7726 138.2 142.5
NCs 6.64 À17.00 À17.26 19.60 24.07 7681 7790 133 134
Nanocrystals-mannitol 6.42 À11.48 À11.24 32.26 21.19 6047 6195 111.6 114.3
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 5
Downloadedby[LIULibraries]at07:0905May2016
mannitol, they showed different wetting behavior in both water
and pH 1.2, resulting in a significantly different redispersion profile.
One possible reason might be the generation of nanocrystalline
particles with a high adhesive hydrophobic surface, which has
more of a tendency to form aggregates in the absence of the dis-
persion agent (mannitol).
Initial dissolution rate determination
The dissolution studies were performed to discriminate the effect-
iveness of the micro suspension, nanosuspension, and nanosuspen-
sion with mannitol upon spray drying. The in-vitro dissolution
profile of a formulation can serve as a predictive tool for in-vivo
analysis40,41
. Here, the energy of the formulation system is reduced
upon spray drying and with the rapid removal of water. For nano-
particles, the rate limiting and more important step is the solvation
step (surface kinetics) at the solid–liquid interface (the dissociation
of drug molecules from the solid)39,42
. Therefore, the
Noyes–Whitney model seems to be ineffective in generating the
discrimination in the dissolution process of the NCs with a mean-
ingful way.
Our objective was to design a conventional in-vitro dissol-
ution method, which can be easily applied to discriminate the
dissolution profiles among the MCs, NCs, and NCm. Initially, the
dissolution studies were carried out by maintaining the sink con-
dition as described in the Method section (n ¼ 3). The sink con-
dition dissolution profile did not show significant discrimination
among MCs, NCs, or NCm (Figure 6(a)). Therefore, for efficient
quantitative discrimination, the dissolution was carried out under
non-sink conditions (n ¼ 3, Figure 6(a) and (b)). The non-sink dis-
solution displayed discrimination in the dissolution profile by
reducing the dissolution velocity. In the sink condition dissol-
ution, the initial dissolution rate ratio of NCs and NCm is negli-
gible. But, as the saturation level increased, the discrimination
increased, and at the 90% saturation level, the initial dissolution
rate ratio for NCs to MCs was 1.8, NCm to MCs was 6.7, and
NCm to NCs was 4.9-fold compared to their dissolution ratios at
initial phase in sink conditions (Table 3). The reported dissolution
rate enhancement behavior can be attributed to the presence of
mannitol during spray drying. This can be further explained by
the improvement in the wetting property and a reduction of
the interfacial tension (tension at solid–liquid interface during
dissolution) for the SD particles of NCm. Also, the partial conver-
sion of crystalline form to amorphous might play a contributing
role in the dissolution behavior for the SD products. However,
since the conversion of crystalline form to amorphous form is
minor (probably less than 5%), the rate of crystallization of the
amorphous form during dissolution expected to be very fast.
Therefore, it will have negligible impact on the dissolution
behavior. The current study justifies the use of non-sink condi-
tion over sink condition for better quantitative discrimination of
nanocrystalline and microcrystalline powder dissolution.
Conclusion
Much of the published work in the area of drying nanocrystalline
suspensions has focused on the impact of the process parameters
on the final product. This study shows the importance of particle
Figure 5. Redispersion study of SD powders of NCs and NCm (a) in pH 1.2, and (b) water.
Figure 6. In-vitro sink and In-vitro non-sink dissolution: comparing MCs, NCs, and NCm: (a) 0% and 20% saturation, and (b) 50% and 90% saturation levels.
6 D. A. SHAH ET AL.
Downloadedby[LIULibraries]at07:0905May2016
size reduction and the incorporation of mannitol as a dispersing
agent during the spray drying of nanocrystalline suspension of
poorly soluble compounds (BCS class II/IV). Upon spray drying, the
fundamental properties (surface and in-vitro dissolution under non-
sink condition) change drastically. The interfacial tension at the
particle moisture interface and redispersion behavior and non-sink
dissolution profile of griseofulvin–mannitol nanocrystalline suspen-
sions show superior performance as compared to the micron/un-
milled and nanocrystalline suspension formulations. The present
study provides an in-depth understanding to a formulation scien-
tist who can quantitate the change in the interfacial tension (at
particle moisture interface) and in the in-vitro dissolution rate
enhancement during initial phase. This provides a platform to
apply a practical approach to screen and rank order the SD formu-
lation with different initial particle size (before drying) with manni-
tol as a dispersing agent.
Disclosure statement
The authors report no declaration of interests.
References
1. Lipinski CA. Drug-like properties and the causes of poor solu-
bility and poor permeability. J Pharmacol Toxicol Methods
2000;44:235–49.
2. Hywel D, Williams NLT, Charman SA, et al. Strategies to
address low drug solubility in discovery and development.
Pharmacol Rev 2013;65:315–499.
3. Shegokar R, Muller RH. Nanocrystals: industrially feasible
multifunctional formulation technology for poorly soluble
actives. Int J Pharm 2010;399:129–39.
4. Serajuddin ATM. Solid dispersion of poorly water-soluble
drugs: early promises, subsequent problems, and recent
breakthroughs. J Pharm Sci 1999;88:1058–66.
5. Dave RH, Shah DA, Patel PG. Development and evaluation of
high loading oral dissolving film of aspirin and acetamino-
phen. J Pharm Sci Pharmacol 2014;1:112–22.
6. Mou D, Chen H, Wan J, et al. Potent dried drug nanosuspen-
sions for oral bioavailability enhancement of poorly soluble
drugs with pH-dependent solubility. Int J Pharm
2011;413:237–44.
7. Merisko-Liversidge E, Liversidge GG, Cooper ER. Nanosizing: a
formulation approach for poorly-water-soluble compounds.
Eur J Pharm Sci 2003;18:113–20.
8. Murdande SB, Shah DA, Dave RH. Impact of nanosizing on
solubility and dissolution rate of poorly soluble pharmaceuti-
cals. J Pharm Sci 2015;104:2094–102.
9. Shah DA, Murdande SB, Dave RH. A review: pharmaceutical
and pharmacokinetic aspect of nanocrystalline suspensions. J
Pharm Sci 2016;105:10–24.
10. Kumar S, Jog R, Shen J, et al. Formulation and performance
of danazol nano-crystalline suspensions and spray dried pow-
ders. Pharm Res 2015;32:1694–703.
11. Song J, Wang Y, Song Y, et al. Development and character-
isation of ursolic acid nanocrystals without stabilizer having
improved dissolution rate and in vitro anticancer activity.
AAPS PharmSciTech 2014;15:11–9.
12. M€uller RH, Peters K. Nanosuspensions for the formulation of
poorly soluble drugs: I. Preparation by a size-reduction tech-
nique. Int J Pharm 1998;160:229–37.
13. Rabinow BE. Nanosuspensions in drug delivery. Nat Rev Drug
Discov 2004;3:785–96.
14. Cooper ER. Nanoparticles: a personal experience for formulat-
ing poorly water soluble drugs. J Control Release
2010;141:300–2.
15. Van Eerdenbrugh B, Vermant J, Martens JA, et al. Solubility
increases associated with crystalline drug nanoparticles:
methodologies and significance. Mol Pharm 2010;7:1858–70.
16. Anhalt K, Geissler S, Harms M, et al. Development of a new
method to assess nanocrystal dissolution based on light scat-
tering. Pharm Res 2012;29:2887–901.
17. Ghosh I, Bose S, Vippagunta R, Harmon F. Nanosuspension
for improving the bioavailability of a poorly soluble drug and
screening of stabilizing agents to inhibit crystal growth. Int J
Pharm 2011;409:260–8.
18. Chaubal MV, Popescu C. Conversion of nanosuspensions into
dry powders by spray drying: a case study. Pharm Res
2008;25:2302–8.
19. Zuo B, Sun Y, Li H, et al. Preparation and in vitro/in vivo evalu-
ation of fenofibrate nanocrystals. Int J Pharm 2013;455:267–75.
20. Isa L, Lucas F, Wepf R, Reimhult E. Measuring single-nanopar-
ticle wetting properties by freeze-fracture shadow-casting
cryo-scanning electron microscopy. Nat Commun 2011;2:438.
doi:10.1038/ncomms1441.
21. Van Eerdenbrugh B, Froyen L, Van Humbeeck J, et al. Drying
of crystalline drug nanosuspensions – the importance of sur-
face hydrophobicity on dissolution behavior upon redisper-
sion. Eur J Pharm Sci 2008;35:127–35.
22. Beck C, Sievens-Figueroa L, G€artner K, et al. Effects of stabil-
izers on particle redispersion and dissolution from polymer
strip films containing liquid antisolvent precipitated griseoful-
vin particles. Powder Technol 2013;236:37–51.
23. Hu J, Ng WK, Dong Y, et al. Continuous and scalable process
for water-redispersible nanoformulation of poorly aqueous
soluble APIs by antisolvent precipitation and spray-drying. Int
J Pharm 2011;404:198–204.
24. Niwa T, Danjo K. Design of self-dispersible dry nanosuspen-
sion through wet milling and spray freeze-drying for poorly
water-soluble drugs. Eur J Pharm Sci 2013;50:272–81.
25. Bojnanska E, Kalina M, Parizek L, et al. Determination of crit-
ical parameters of drug substance influencing dissolution: a
case study. BioMed Res Int 2014;2014. doi:http://dx.doi.org/
10.1155/2014/929248.
Table 3. Quantitation of initial dissolution rate ratios.
Conditions
Dissolved in 2 min (%) Initial dissolution rate ratios
MC NC Nanocrystals-mannitol NCa
/MCb
NCmc
/MC NCm/NC
Sink 31 83 96 2.7 3.1 1.1
20% 11 33 37 3.0 3.3 1.1
50% 6 13 30 2.16 5.0 2.3
90% 4 7 28 1.75 7.0 4.0
a
NC spray-dried, b
micro SD powder, c
nanocrystal with mannitol spray-dried.
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 7
Downloadedby[LIULibraries]at07:0905May2016
26. Sahoo NG, Kakran M, Shaal LA, et al. Preparation and charac-
terization of quercetin nanocrystals. J Pharm Sci 2011;
100:2379–90.
27. Okoye PC. To systemically characterize magnesium stearate
polymorphs and practically evaluate their effect on physico-
chemical properties of naproxen (BCS Class II) and acet-
aminophen (BCS Class III) as model drugs. Edited by
Pharmaceutical Sciences. Brooklyn (NY): Long Island
University;2013:190.
28. Prajapati H, Dalrymple DM, Serajuddin AT. In vitro dispersion
test can serve as a predictive method for assessing perform-
ance of lipid-based drug delivery systems. J Excipient Food
Chem 2013;4:111–25.
29. Sun J, Wang F, Sui Y, et al. Effect of particle size on solubility,
dissolution rate, and oral bioavailability: evaluation using
coenzyme Q(10) as naked nanocrystals. Int J Nanomed
2012;7:5733–44.
30. Chidambaram N, Burgess DJ. A novel in vitro release method
for submicron-sized dispersed systems. AAPS PharmSci
1999;1:32–40
31. Kocbek P, Baumgartner S, Kristl J. Preparation and evaluation
of nanosuspensions for enhancing the dissolution of poorly
soluble drugs. Int J Pharm 2006;312:179–86.
32. Jinno J, Kamada N, Miyake M, et al. Effect of particle size
reduction on dissolution and oral absorption of a poorly
water-soluble drug, cilostazol, in beagle dogs. J Control
Release 2006;111:56–64.
33. Ma Q, Sun H, Che E, et al. Uniform nano-sized valsartan for
dissolution and bioavailability enhancement: Influence of par-
ticle size and crystalline state. Int J Pharm 2013;441:75–81.
34. Liversidge GG, Cundy KC. Particle size reduction for improve-
ment of oral bioavailability of hydrophobic drugs: I. Absolute
oral bioavailability of nanocrystalline danazol in beagle dogs.
Int J Pharm 1995;125:91–7.
35. Korang-Yeboah M, Rahman Z, Shah D, et al. Impact of formu-
lation and process variables on solid-state stability of theo-
phylline in controlled release formulations. Int J Pharm
2016;499:20–8.
36. Wu W, Nancollas G. A new understanding of the relationship
between solubility and particle size. J Solut Chem 1998;
27:521–31.
37. Kaur J, Aggarwal G, Singh G, Rana AC. Improvement of drug
soluility using solid dispersion. Int J Pharm Pharm Sci
2012;4:47–53
38. Li X, Vogt FG, Hayes D, Mansour HM. Design, characteriza-
tion, and aerosol dispersion performance modeling of
advanced spray-dried microparticulate/nanoparticulate man-
nitol powders for targeted pulmonary delivery as dry powder
inhalers. J Aeros Med Pulm Drug Deliv 2014;27:81–93.
39. Crisp MT, Tucker CJ, Rogers TL, et al. Turbidimetric measure-
ment and prediction of dissolution rates of poorly soluble
drug nanocrystals. J Control Release 2007;117:351–9.
40. Heng D, Cutler DJ, Chan HK, et al. What is a suitable dissol-
ution method for drug nanoparticles? Pharm Res
2008;25:1696–701.
41. Kataoka M, Itsubata S, Masaoka Y, et al. In vitro dissolution/
permeation system to predict the oral absorption of poorly
water-soluble drugs: effect of food and dose strength on it.
Biol Pharm Bull 2011;34:401–7.
42. Shekunov BY, Chattopadhyay P, Seitzinger J, Huff R.
Nanoparticles of poorly water-soluble drugs prepared by
supercritical fluid extraction of emulsions. Pharm Res
2006;23:196–204.
Appendix A
The adsorption of moisture onto multilayer surfaces is believed to
follow the Brunauer–Emmett–Teller (BET) model. Similarly, we make
the assumption that adsorption of moisture onto spray-dried pow-
der particles follows the BET multilayer model. In this assignment,
an attempt is made to relate surface energy from BET to interfacial
tension based on moisture adsorption onto powder particles.
Determination of BET parameters
The BET equation is given by:
m
m0
¼
CX
ð1 À XÞ½1 þ ðC À 1ÞXŠ
(A1)
From thermodynamics, the ratio of partial pressure of water
and that of pure substance is represented by:
aw ¼
p
p0
¼ X (A2)
We can now rearrange Equation (A1) to obtain the multilayer
model of BET as shown by:
m ¼
awm0C
ð1 À awÞ½1 þ awðC À 1ÞŠ
(A3)
where m is the moisture content in g/g solids; aw, is water
activity; m0, is the moisture content of monolayer in g/g solids;
C ¼ e
DE = RT
, the BET model is typically applicable up to 0.5aw.
The experimental application of the BET multilayer model involves
obtaining some initial values of water activity, aw, with correspond-
ing moisture content values, m. The analytical method involves lin-
earization of Equation (A3) to obtain:
½1 þ awðc À 1ÞŠ
m0c
¼
aw
ð1 À awÞm
(A4)
Finally,
aw
ð1 À awÞm
¼ aw
c À 1
m0c
 
þ
1
m0c
(A5)
where a plot of aw
ð1Àaw Þm versus aw gives a straight line with slope as
cÀ1
m0c
h i
and the intercept as 1
m0c. The BET parameters, m0, and C were
experimentally obtained as m0 ¼ 1
slopeþintercept and C ¼ 1
interceptÃm0
.
Determination of the cross-sectional area of the water molecule
Assume monolayer saturable sites with each molecule of water
spreading across the spray-dried powder particle. From quantum
thermodynamics, the spatial separation between two water mole-
cules is given by:
V
N
 1=3
18x10À6
m3
6x1023
 1=3
¼ ð30x10À30
m3
Þ1=3
¼ 3x10À10
m (A6)
8 D. A. SHAH ET AL.
Downloadedby[LIULibraries]at07:0905May2016
3x10À10
m à 3x10À10
m ¼ 9x10À20
m2
¼ 0:9x10À19
m2
The cross-sectional area of a water molecule is then derived as:
Note: The actual cross-sectional area of a water molecule:
1.06 Â 10À19
m2
.
Determination of interfacial tension (c) between particle and
moisture
The molecular relationship between solid–liquid interface may be
described by the changes in surface energy (DE). The deposition of
monolayer of moisture on the powder molecules creates an inter-
facial tension that is expressed based on Young–Laplace expression:
DE ¼ cA
where DE is the surface or binding energy of the monolayer; c is
the molecular interfacial tension; and A is the molecular cross sec-
tional area.
If we assume non-interactive particle–particle behavior as
espoused by BET, then the molecular interfacial tension between
powder and water molecules could be estimated from the binding
or surface energy at the saturable sites as given by:
c ¼
DE
NAvogadro
Ã
1
AH20
(A7)
where DE is monolayer binding or surface energy; NAvogadro is the
Avogadro’s number,
(6:02x1023
molecules=mol); AH20 is the cross-sectional area of
water molecule (1:06x10À19
m2
). In conclusion, the BET model
allowed the estimation of powder-moisture interfacial tension
through the construct of surface energy of saturable
monolayer.
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 9
Downloadedby[LIULibraries]at07:0905May2016

More Related Content

What's hot

Problems of variable
Problems of variableProblems of variable
Problems of variableSAKSHI YADAV
 
Dissolution Enhancement of BCS Class 4 Dssrugs Using Quality by Design Approa...
Dissolution Enhancement of BCS Class 4 Dssrugs Using Quality by Design Approa...Dissolution Enhancement of BCS Class 4 Dssrugs Using Quality by Design Approa...
Dissolution Enhancement of BCS Class 4 Dssrugs Using Quality by Design Approa...inventionjournals
 
Dissolution by Dr. Neeraj Mishra professor pharmaceutics
Dissolution by Dr. Neeraj Mishra professor pharmaceuticsDissolution by Dr. Neeraj Mishra professor pharmaceutics
Dissolution by Dr. Neeraj Mishra professor pharmaceuticsNeeraj Mishra
 
Pensee design and evaluation of ramipril 1
Pensee design and evaluation of ramipril 1Pensee design and evaluation of ramipril 1
Pensee design and evaluation of ramipril 1sonalsuryawanshi2
 
Liquisolid technique as a tool for enhancement
Liquisolid technique as a tool for enhancementLiquisolid technique as a tool for enhancement
Liquisolid technique as a tool for enhancementMeghraj Suryawanshi (PDCR)
 
Sagar Goda dissolution studies
Sagar Goda dissolution studies Sagar Goda dissolution studies
Sagar Goda dissolution studies Sagar Goda
 
DISSOLUTION PARAMETERS AND ITS APPARATUS
DISSOLUTION PARAMETERS AND ITS APPARATUSDISSOLUTION PARAMETERS AND ITS APPARATUS
DISSOLUTION PARAMETERS AND ITS APPARATUSROHIT
 
Formulation and Evaluation of Liquisolid Compact of Etoricoxib for Solubility...
Formulation and Evaluation of Liquisolid Compact of Etoricoxib for Solubility...Formulation and Evaluation of Liquisolid Compact of Etoricoxib for Solubility...
Formulation and Evaluation of Liquisolid Compact of Etoricoxib for Solubility...ijpsmjournal
 
Application of preformulation consideration in the development of
Application of preformulation consideration in the development ofApplication of preformulation consideration in the development of
Application of preformulation consideration in the development ofArpan Dhungel
 
factors affecting dissolution rate a full view.
 factors affecting dissolution rate a full view. factors affecting dissolution rate a full view.
factors affecting dissolution rate a full view.aishwaryashiremath
 
Compendial methods of dissolution
Compendial methods of dissolutionCompendial methods of dissolution
Compendial methods of dissolutionHemanth KG
 
Enhancement of Solubility By Solid Dispersion- Presented By Mr.Ajinkya Nikam...
 Enhancement of Solubility By Solid Dispersion- Presented By Mr.Ajinkya Nikam... Enhancement of Solubility By Solid Dispersion- Presented By Mr.Ajinkya Nikam...
Enhancement of Solubility By Solid Dispersion- Presented By Mr.Ajinkya Nikam...Ajinkya Nikam
 
Biopharmaceutic considerations in drug product design and in
Biopharmaceutic considerations in drug product design and inBiopharmaceutic considerations in drug product design and in
Biopharmaceutic considerations in drug product design and inSUJITHA MARY
 
Theories of drug dissolution Biopharmaceutics
Theories of drug dissolution BiopharmaceuticsTheories of drug dissolution Biopharmaceutics
Theories of drug dissolution BiopharmaceuticsHarshvardhanRaval1
 
solid dispersion-polymorphism
solid dispersion-polymorphismsolid dispersion-polymorphism
solid dispersion-polymorphismGaurav Kr
 
Dissolution of Solid dosages form
Dissolution of Solid dosages formDissolution of Solid dosages form
Dissolution of Solid dosages formMd. Mizanur Rahman
 
Application of preformulation_consideration_in_the_development_of
Application of preformulation_consideration_in_the_development_ofApplication of preformulation_consideration_in_the_development_of
Application of preformulation_consideration_in_the_development_ofSUJIT DAS
 

What's hot (20)

Problems of variable
Problems of variableProblems of variable
Problems of variable
 
Dissolution Enhancement of BCS Class 4 Dssrugs Using Quality by Design Approa...
Dissolution Enhancement of BCS Class 4 Dssrugs Using Quality by Design Approa...Dissolution Enhancement of BCS Class 4 Dssrugs Using Quality by Design Approa...
Dissolution Enhancement of BCS Class 4 Dssrugs Using Quality by Design Approa...
 
Dissolution by Dr. Neeraj Mishra professor pharmaceutics
Dissolution by Dr. Neeraj Mishra professor pharmaceuticsDissolution by Dr. Neeraj Mishra professor pharmaceutics
Dissolution by Dr. Neeraj Mishra professor pharmaceutics
 
Pensee design and evaluation of ramipril 1
Pensee design and evaluation of ramipril 1Pensee design and evaluation of ramipril 1
Pensee design and evaluation of ramipril 1
 
Liquisolid technique as a tool for enhancement
Liquisolid technique as a tool for enhancementLiquisolid technique as a tool for enhancement
Liquisolid technique as a tool for enhancement
 
Sagar Goda dissolution studies
Sagar Goda dissolution studies Sagar Goda dissolution studies
Sagar Goda dissolution studies
 
DISSOLUTION PARAMETERS AND ITS APPARATUS
DISSOLUTION PARAMETERS AND ITS APPARATUSDISSOLUTION PARAMETERS AND ITS APPARATUS
DISSOLUTION PARAMETERS AND ITS APPARATUS
 
Formulation and Evaluation of Liquisolid Compact of Etoricoxib for Solubility...
Formulation and Evaluation of Liquisolid Compact of Etoricoxib for Solubility...Formulation and Evaluation of Liquisolid Compact of Etoricoxib for Solubility...
Formulation and Evaluation of Liquisolid Compact of Etoricoxib for Solubility...
 
Application of preformulation consideration in the development of
Application of preformulation consideration in the development ofApplication of preformulation consideration in the development of
Application of preformulation consideration in the development of
 
factors affecting dissolution rate a full view.
 factors affecting dissolution rate a full view. factors affecting dissolution rate a full view.
factors affecting dissolution rate a full view.
 
Apt lab manual
Apt lab manualApt lab manual
Apt lab manual
 
Compendial methods of dissolution
Compendial methods of dissolutionCompendial methods of dissolution
Compendial methods of dissolution
 
Enhancement of Solubility By Solid Dispersion- Presented By Mr.Ajinkya Nikam...
 Enhancement of Solubility By Solid Dispersion- Presented By Mr.Ajinkya Nikam... Enhancement of Solubility By Solid Dispersion- Presented By Mr.Ajinkya Nikam...
Enhancement of Solubility By Solid Dispersion- Presented By Mr.Ajinkya Nikam...
 
Biopharmaceutic considerations in drug product design and in
Biopharmaceutic considerations in drug product design and inBiopharmaceutic considerations in drug product design and in
Biopharmaceutic considerations in drug product design and in
 
Theories of drug dissolution Biopharmaceutics
Theories of drug dissolution BiopharmaceuticsTheories of drug dissolution Biopharmaceutics
Theories of drug dissolution Biopharmaceutics
 
Pre formulaton
Pre formulatonPre formulaton
Pre formulaton
 
Bhavani sem
Bhavani semBhavani sem
Bhavani sem
 
solid dispersion-polymorphism
solid dispersion-polymorphismsolid dispersion-polymorphism
solid dispersion-polymorphism
 
Dissolution of Solid dosages form
Dissolution of Solid dosages formDissolution of Solid dosages form
Dissolution of Solid dosages form
 
Application of preformulation_consideration_in_the_development_of
Application of preformulation_consideration_in_the_development_ofApplication of preformulation_consideration_in_the_development_of
Application of preformulation_consideration_in_the_development_of
 

Viewers also liked

A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...Dhaval shah
 
Fundamentals of spray drying
Fundamentals of spray dryingFundamentals of spray drying
Fundamentals of spray dryingDowntp Livros
 
spray dried
spray driedspray dried
spray driedsecub
 
Spray Drying equipment
Spray Drying equipmentSpray Drying equipment
Spray Drying equipmentnjsfspraydryer
 
Scott Ellis Ai Ch E 2010 Annual Meeting Qb D Strategy
Scott Ellis Ai Ch E 2010 Annual Meeting Qb D StrategyScott Ellis Ai Ch E 2010 Annual Meeting Qb D Strategy
Scott Ellis Ai Ch E 2010 Annual Meeting Qb D Strategyruggles62
 
Tour10 venezuela - navarro
Tour10   venezuela - navarroTour10   venezuela - navarro
Tour10 venezuela - navarropaulvillanueva01
 
Soalan bm t4 kertas 2
Soalan bm  t4  kertas 2Soalan bm  t4  kertas 2
Soalan bm t4 kertas 2fieza848
 
LAS DROGAS
LAS DROGASLAS DROGAS
LAS DROGASbryan_99
 
Catalogo Creazione d'Impresa - Biella 2016
Catalogo Creazione d'Impresa - Biella 2016Catalogo Creazione d'Impresa - Biella 2016
Catalogo Creazione d'Impresa - Biella 2016Clara Crosa Galant
 
User Test Introduction
User Test IntroductionUser Test Introduction
User Test IntroductionErgonline
 
5 клас урок 7
5 клас урок 75 клас урок 7
5 клас урок 7Iren50
 
Инструкция сигвеи Q7 Q6
Инструкция сигвеи Q7 Q6Инструкция сигвеи Q7 Q6
Инструкция сигвеи Q7 Q6Сергей К.
 

Viewers also liked (16)

A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
A Review- Pharmaceutical and Pharmacokinetic Aspect of Nanocrystalline Suspen...
 
Fundamentals of spray drying
Fundamentals of spray dryingFundamentals of spray drying
Fundamentals of spray drying
 
spray dried
spray driedspray dried
spray dried
 
Spray Drying equipment
Spray Drying equipmentSpray Drying equipment
Spray Drying equipment
 
Scott Ellis Ai Ch E 2010 Annual Meeting Qb D Strategy
Scott Ellis Ai Ch E 2010 Annual Meeting Qb D StrategyScott Ellis Ai Ch E 2010 Annual Meeting Qb D Strategy
Scott Ellis Ai Ch E 2010 Annual Meeting Qb D Strategy
 
Spray Dryer
Spray DryerSpray Dryer
Spray Dryer
 
Spray Drying
Spray DryingSpray Drying
Spray Drying
 
spray drying
spray dryingspray drying
spray drying
 
Tour10 venezuela - navarro
Tour10   venezuela - navarroTour10   venezuela - navarro
Tour10 venezuela - navarro
 
Soalan bm t4 kertas 2
Soalan bm  t4  kertas 2Soalan bm  t4  kertas 2
Soalan bm t4 kertas 2
 
LAS DROGAS
LAS DROGASLAS DROGAS
LAS DROGAS
 
Catalogo Creazione d'Impresa - Biella 2016
Catalogo Creazione d'Impresa - Biella 2016Catalogo Creazione d'Impresa - Biella 2016
Catalogo Creazione d'Impresa - Biella 2016
 
Tema 8 La Edad Media
Tema 8 La Edad MediaTema 8 La Edad Media
Tema 8 La Edad Media
 
User Test Introduction
User Test IntroductionUser Test Introduction
User Test Introduction
 
5 клас урок 7
5 клас урок 75 клас урок 7
5 клас урок 7
 
Инструкция сигвеи Q7 Q6
Инструкция сигвеи Q7 Q6Инструкция сигвеи Q7 Q6
Инструкция сигвеи Q7 Q6
 

Similar to Influence of spray drying and dispersing agent on surface and dissolution properties of griseofulvin micro and nanocrystals

Effect of diluent types and soluble diluents particle size on the dissolution...
Effect of diluent types and soluble diluents particle size on the dissolution...Effect of diluent types and soluble diluents particle size on the dissolution...
Effect of diluent types and soluble diluents particle size on the dissolution...Valentyn Mohylyuk
 
Improved Wetting For Improved Solubility and Dissolution of Candesartan Cil...
	 Improved Wetting For Improved Solubility and Dissolution of Candesartan Cil...	 Improved Wetting For Improved Solubility and Dissolution of Candesartan Cil...
Improved Wetting For Improved Solubility and Dissolution of Candesartan Cil...iosrphr_editor
 
Bioavailability of Gold nanoparticles.pptx
Bioavailability of Gold nanoparticles.pptxBioavailability of Gold nanoparticles.pptx
Bioavailability of Gold nanoparticles.pptxImdad H. Mukeri
 
Preformulation/pharmaceutical preformulation
Preformulation/pharmaceutical preformulationPreformulation/pharmaceutical preformulation
Preformulation/pharmaceutical preformulationM R S
 
Effect of aqueous suspensions of titanium dioxide in photoreactor with
Effect of aqueous suspensions of titanium dioxide in photoreactor withEffect of aqueous suspensions of titanium dioxide in photoreactor with
Effect of aqueous suspensions of titanium dioxide in photoreactor withAmeer Al-Ameedee
 
Tapioca starch-pullulan interaction during gelation and retrogradation
Tapioca starch-pullulan interaction during gelation and retrogradationTapioca starch-pullulan interaction during gelation and retrogradation
Tapioca starch-pullulan interaction during gelation and retrogradationMostafa Gouda
 
Phenhylephrine Hydrocloride Gastro Retentive Floating Matrix Tablets Design a...
Phenhylephrine Hydrocloride Gastro Retentive Floating Matrix Tablets Design a...Phenhylephrine Hydrocloride Gastro Retentive Floating Matrix Tablets Design a...
Phenhylephrine Hydrocloride Gastro Retentive Floating Matrix Tablets Design a...ijtsrd
 
Formulation Development and Evaluation of Mouth Dissolving Film of Ziprasidon...
Formulation Development and Evaluation of Mouth Dissolving Film of Ziprasidon...Formulation Development and Evaluation of Mouth Dissolving Film of Ziprasidon...
Formulation Development and Evaluation of Mouth Dissolving Film of Ziprasidon...ijtsrd
 
Theories of solubulisation
Theories of solubulisationTheories of solubulisation
Theories of solubulisationvenkatesh thota
 
Nanosuspension 160410194151
Nanosuspension 160410194151Nanosuspension 160410194151
Nanosuspension 160410194151Shubham Dhakne
 
Formulation and Development of Modified Release Biphasic Compressed Tablet of...
Formulation and Development of Modified Release Biphasic Compressed Tablet of...Formulation and Development of Modified Release Biphasic Compressed Tablet of...
Formulation and Development of Modified Release Biphasic Compressed Tablet of...ijtsrd
 

Similar to Influence of spray drying and dispersing agent on surface and dissolution properties of griseofulvin micro and nanocrystals (20)

Effect of diluent types and soluble diluents particle size on the dissolution...
Effect of diluent types and soluble diluents particle size on the dissolution...Effect of diluent types and soluble diluents particle size on the dissolution...
Effect of diluent types and soluble diluents particle size on the dissolution...
 
dissolution
dissolutiondissolution
dissolution
 
Poster presentation
Poster presentationPoster presentation
Poster presentation
 
self microemulsified bosentan systems
self microemulsified bosentan systemsself microemulsified bosentan systems
self microemulsified bosentan systems
 
Improved Wetting For Improved Solubility and Dissolution of Candesartan Cil...
	 Improved Wetting For Improved Solubility and Dissolution of Candesartan Cil...	 Improved Wetting For Improved Solubility and Dissolution of Candesartan Cil...
Improved Wetting For Improved Solubility and Dissolution of Candesartan Cil...
 
Swapnil nanosuspension ppt
Swapnil nanosuspension pptSwapnil nanosuspension ppt
Swapnil nanosuspension ppt
 
Pre-Formulation
Pre-FormulationPre-Formulation
Pre-Formulation
 
Bioavailability of Gold nanoparticles.pptx
Bioavailability of Gold nanoparticles.pptxBioavailability of Gold nanoparticles.pptx
Bioavailability of Gold nanoparticles.pptx
 
Preformulation/pharmaceutical preformulation
Preformulation/pharmaceutical preformulationPreformulation/pharmaceutical preformulation
Preformulation/pharmaceutical preformulation
 
Nanosuspension
Nanosuspension Nanosuspension
Nanosuspension
 
Effect of aqueous suspensions of titanium dioxide in photoreactor with
Effect of aqueous suspensions of titanium dioxide in photoreactor withEffect of aqueous suspensions of titanium dioxide in photoreactor with
Effect of aqueous suspensions of titanium dioxide in photoreactor with
 
Tapioca starch-pullulan interaction during gelation and retrogradation
Tapioca starch-pullulan interaction during gelation and retrogradationTapioca starch-pullulan interaction during gelation and retrogradation
Tapioca starch-pullulan interaction during gelation and retrogradation
 
Nanosuspension
NanosuspensionNanosuspension
Nanosuspension
 
Phenhylephrine Hydrocloride Gastro Retentive Floating Matrix Tablets Design a...
Phenhylephrine Hydrocloride Gastro Retentive Floating Matrix Tablets Design a...Phenhylephrine Hydrocloride Gastro Retentive Floating Matrix Tablets Design a...
Phenhylephrine Hydrocloride Gastro Retentive Floating Matrix Tablets Design a...
 
Formulation Development and Evaluation of Mouth Dissolving Film of Ziprasidon...
Formulation Development and Evaluation of Mouth Dissolving Film of Ziprasidon...Formulation Development and Evaluation of Mouth Dissolving Film of Ziprasidon...
Formulation Development and Evaluation of Mouth Dissolving Film of Ziprasidon...
 
Theories of solubulisation
Theories of solubulisationTheories of solubulisation
Theories of solubulisation
 
Nanosuspension
NanosuspensionNanosuspension
Nanosuspension
 
Nanosuspension 160410194151
Nanosuspension 160410194151Nanosuspension 160410194151
Nanosuspension 160410194151
 
Formulation and Development of Modified Release Biphasic Compressed Tablet of...
Formulation and Development of Modified Release Biphasic Compressed Tablet of...Formulation and Development of Modified Release Biphasic Compressed Tablet of...
Formulation and Development of Modified Release Biphasic Compressed Tablet of...
 
Solid lipid nanoparticles
Solid lipid nanoparticles Solid lipid nanoparticles
Solid lipid nanoparticles
 

Influence of spray drying and dispersing agent on surface and dissolution properties of griseofulvin micro and nanocrystals

  • 1. Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=iddi20 Download by: [LIU Libraries] Date: 05 May 2016, At: 07:09 Drug Development and Industrial Pharmacy ISSN: 0363-9045 (Print) 1520-5762 (Online) Journal homepage: http://www.tandfonline.com/loi/iddi20 Influence of spray drying and dispersing agent on surface and dissolution properties of griseofulvin micro and nanocrystals Dhaval A. Shah, Manan Patel, Sharad B. Murdande & Rutesh H. Dave To cite this article: Dhaval A. Shah, Manan Patel, Sharad B. Murdande & Rutesh H. Dave (2016): Influence of spray drying and dispersing agent on surface and dissolution properties of griseofulvin micro and nanocrystals, Drug Development and Industrial Pharmacy, DOI: 10.1080/03639045.2016.1178770 To link to this article: http://dx.doi.org/10.1080/03639045.2016.1178770 Accepted author version posted online: 14 Apr 2016. Published online: 04 May 2016. Submit your article to this journal Article views: 13 View related articles View Crossmark data
  • 2. RESEARCH ARTICLE Influence of spray drying and dispersing agent on surface and dissolution properties of griseofulvin micro and nanocrystals Dhaval A. Shaha , Manan Patela , Sharad B. Murdandeb and Rutesh H. Davea a Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA; b Drug Product Design, Pfizer Worldwide R&D, Groton, CT, USA ABSTRACT The purpose for the current research is to compare and evaluate physiochemical properties of spray-dried (SD) microcrystals (MCs), nanocrystals (NCs), and nanocrystals with a dispersion agent (NCm) from a poorly soluble compound. The characterization was carried out by performing size and surface analysis, interfacial tension (at particle moisture interface), and in-vitro drug dissolution rate experiments. Nanosuspensions were prepared by media milling and were spray-dried. The SD powders that were obtained were character- ized morphologically using scanning electron microscopy (SEM), polarized light microscopy (PLM), and Flowchem. Solid-state characterization was performed using X-ray powder diffraction (XRPD), Fourier trans- fer infrared spectroscopy (FT-IR), and differential scanning calorimetry (DSC) for the identification of the crystalline nature of all the SD powders. The powders were characterized for their redispersion tendency in the water and in pH 1.2. Significant differences in redispersion were noted for both the NCs in both dissol- ution media. The interfacial tension for particle moisture interface was determined by applying the BET (Braunauer–Emmett–Teller) equation to the vapor sorption data. No significant reduction in the interfacial tension was observed between MCs and NCs; however, a significant reduction in the interfacial tension was observed for NCm at both 25 C and 35 C temperatures. The difference in interfacial tension and redisper- sion behavior can be attributed to a difference in the wetting tendency for all the SD powders. The dissol- ution studies were carried out under sink and under non-sink conditions. The non-sink dissolution approach was found suitable for quantification of the dissolution rate enhancement, and also for providing the rank order to the SD formulations. ARTICLE HISTORY Received 29 December 2015 Revised 30 March 2016 Accepted 7 April 2016 Published online 3 May 2016 KEYWORDS Griseofulvin; interfacial tension; nanocrystals; non- sink dissolution; redispersion; spray dryer Introduction It has been reported that more than half of the identified potential drug candidates have challenges related to aqueous solubility and/ or the dissolution rate1 . A majority of these compounds belong to the BCS class II and IV categories2,3 . The rate and the extent of absorption of the Class II compounds are highly dependent on the performance of the formulated product4–6 . Several formulation approaches have been suggested and reported to overcome these problems. The formulation of nanocrystals (NCs; suspension or solid) is one of the preferred approaches that has been used for the drugs which belong to the above BCS classes7,8 . NCs offer sev- eral advantages, including enhancement in bioavailability, reduc- tion in therapeutic dose, and elimination of the food effect9–14 . The solubility behavior of crystalline nanoparticles can be explained by the Ostwald–Frendulich equation. Based on the pub- lished reports, the thermodynamic solubility enhancement of nano- crystalline particles should be approximately 10–15%8,15,16 . On the other hand, nanosuspensions have a very high surface-to-volume ratio and high Gibbs energy, which indicates the instability of the system. They tend to form aggregates due to Ostwald’s ripen- ing13,17 . Nanosuspensions can be stabilized and their shelf life can be enhanced by converting them into solid powders by spray dry- ing and/or freeze drying10,18 . Upon solidification, the surface properties of dried particles (i.e. size, shape) may tend to change significantly, which will have a dir- ect impact on the wetting and dissolution performance of the formulation19 . Hence, it is crucial to assess the surface and the interfacial properties of the dried particles20,21 . It is also known that these suspensions (micro/nano) may form reversible or nonreversi- ble aggregates upon solidification. Therefore, aggregation is another key product quality parameter that needs to be assessed for effectiveness with the dried particles22,23 . During spray drying, the particle size distribution is governed by the nozzle size24 . It is possible to have different dissolution behavior for formulations with the same particle size due to change in surface wetting prop- erty25 . Therefore, it will be important to determine the role of the interfacial tension (at particle to moisture interface) in governing the redispersion and the kinetic solubility (initial dissolution rate) of NCs and micro crystals (MCs). In the current study, we have formulated spray-dried (SD) nano- crystalline powder (with and without the bulking agent mannitol) and MCs of a poorly soluble drug. Griseofulvin (a BCS Class II com- pound) was selected as a model compound and docusate sodium (ionic stabilizer) and HPC-SL (Hydroxypropyl cellulose-low viscosity grade; polymeric stabilizer) were utilized to prevent agglomeration of nanosuspensions before spray drying, while the mannitol was used as a dispersing agent for spray drying. The SD powders were evaluated for surface characterization (size and shape analysis), solid-state characterization (XRPD, DSC, and FT-IR), interfacial ten- sion (DVSO), redispersion, and in-vitro dissolution rates (sink and non-sink). This study shows the effect of interfacial tension in gov- erning the dissolution of NCs and quantitates the dissolution rate CONTACT Rutesh H. Dave rutesh.dave@liu.edu Arnold Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA. ß 2016 Informa UK Limited, trading as Taylor Francis Group DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY, 2016 http://dx.doi.org/10.1080/03639045.2016.1178770 Downloadedby[LIULibraries]at07:0905May2016
  • 3. enhancement for the SD powders using non-sink conditions to provide them in rank order based on their in-vitro performance. Materials Griseofulvin was obtained from Spectrum Chemicals (New Brunswick, NJ). The yttrium-stabilized zirconium oxide beads were obtained from Glen Mills Inc. (Clifton, NJ). Hydroxypropylcellulose (HPC)–SL was obtained from Nisso America Inc. (New York, NY). Other laboratory chemicals, such as mannitol, docusate sodium USP, acetonitrile, and trifluoroacetic acid (TFA), were purchased from Spectrum Chemicals (New Brunswick, NJ) and were of analyt- ical grades. All the excipients utilized in this study were of high purity. The chemical structure of API and excipients are presented in Figure 1. Methods Wet milling Griseofulvin (50 mg/mL) was suspended in an aqueous stabilizer solution which is a mixture of 1.25% (w/v) HPC–SL (stearic stabil- izer) and 0.05% (w/v) docusate sodium (ionic stabilizer). The sus- pension (500 mL) was milled using the yttrium stabilized zirconium oxide beads (YTZ beads) at a milling speed of 1000 rpm in the con- tinuous model8 . The suspension formulation was milled for 4 h, and the particle size was measured during and following the mill- ing process. The temperature of the sample was maintained below 25 C using the cold water that surrounded the milling chamber to compensate for the heat generated by high-speed milling. The sus- pension was harvested by filtration through a coarse sintered-glass funnel filter (75 mm). The nanosuspension was divided into two equal parts, and in one part the mannitol was dissolved in a 1:2 ratio (Griseofulvin: mannitol). The suspension was then stirred for at least 30 min and then prepared for spray drying. The microcrys- talline suspension was prepared using the same method and the same stabilizer concentration with the absence of the milling vehicle (i.e. YTZ beads). Spray drying Nano (with and without the mannitol) and microcrystalline suspen- sions were spray-dried using a B-290 spray dryer (Buchi Labortechhnik AG, Flawil, Switzerland). The spray dryer was equili- brated using distilled water at 128 C inlet temperature, 3 mL/min feed rate, and 100% aspiration rate. The outlet temperature was approximately 75 C. The aforementioned SD parameters were determined from some preliminary experiments. Once the spray dryer was equilibrated, the suspension formulations were subjected to spray drying with continuous stirring using a magnetic stirrer. SD powders were collected from the collection chamber and immediately analyzed for particle size, process yield, and crystallinity. Particle size, surface and shape analysis A dynamic light scattering (DLS) instrument (Malvern Instruments Inc., Malvern, UK) was used to measure particle size and the size distribution of suspensions. All three SD powders were subjected to particle size and shape analysis. Particle size measurements of SD powders were performed using a Sympatec GmbH System- Partikel-Technik (Sympatec Inc., Pennington, NJ). Briefly, around 100 mg of a sample was subjected for analysis. Compressed air was used to disperse the powder, which is blown through the laser beam and collected by suction after the analysis. Polarized light microscopy (PLM; Model BX60FS; Olympus BX60, Center Valley, PA) and scanning electron microscopy (SEM, ASPEX Scanning Electron Microscope model PSEM 3025 SEM/EDS, Fei Inc., Hillsboro, OR) were conducted to observe the surface morphology and texture of the pure materials and SD powders. The polarized microscopic images were taken using a camera from Diagnostic Instruments Inc. (Sterling Heights, MI), and the analysis was performed with the help of SPOTTM imaging advanced software. For the SEM imaging, the samples were mounted on double-sided carbon tape. The low vacuum (LV) mode was used to prevent the samples from charg- ing. The energy beam and filament drive were adjusted to 20 kev and 72%, respectively. The samples were analyzed under approxi- mately 60% contrast and 30% brightness in secondary electron mode. Particle shape and further morphological characterization were performed using the flow cam machine (Fluid Imaging Technologies, Scarborough, ME). Here, the sample was prepared by dispersing the SD powders into the water and then subjected to analysis. The machine captures particle images at up to 22 frames per second. The analysis was performed by keeping the flow rate at 0.3 mL/min and viewed under 4Â optical lens. The morpho- logical measurements (i.e. shape) of particles were calculated using the Visual spread sheetVR Particle analysis software. Differential scanning calorimetry Modulated differential scanning calorimetric (mDSC) analysis was performed using a Q200 (TA Instruments, New Castle, DE) equipped with a cooling system. A sample (5–10 mg) was weighed into an aluminum pan with a pin hole and hermitically sealed. The DSC parameters used were: a sample equilibration at 30 C, modu- lated for ±0.318 C every 60 s, isothermal for 5 min and heated to 230 C with a ramp of 2 C/min. The DSC cell was purged with nitrogen gas at a flow rate of 50 mL/min. The data obtained were analyzed with the Universal Analysis Software (TA Instruments, New Castle, DE). Powder X-ray diffraction Powder X-ray diffraction was done using a scanning diffractometer (Advanced Diffraction System, Scintag Inc. Model XI, Cupertino, CA), controlled by a computer with diffraction management system software for Windows NT. The radiation used was generated by a copper Ka filter, with a wavelength of 1.54 A at 45 kV and 40 mA. High energy mixture patterns obtained by the X-ray were Figure 1. Chemical structures of a model compound and excipients. 2 D. A. SHAH ET AL. Downloadedby[LIULibraries]at07:0905May2016
  • 4. compared to a reference drug to confirm crystallinity26 . Samples were scanned over a range of 5–40 degrees, using a scan rate of 1 /min and a step size of 0.02. X-ray diffraction patterns were obtained for pure griseofulvin, excipients, physical mixtures, and formulated SD powders. Data analysis for recorded patterns was done using X’Pert Data Viewer software. Infrared spectroscopy (FT-IR) FTIR spectra for all the abovementioned samples were obtained by Nicolet-FTIR equipped with smart ATR (Thermo Fisher Scientific Inc. Madison, WI), and a DTGS-KBr (Deuterated triglycine sulfate) detector was used for background and baseline corrections. Small amounts of the sample were placed on the diamond crystal of the ATR and then the ATR knob was closed. Finally, the spectra were collected from 400 to 4000 cmÀ1 with 264 accumulations at a reso- lution of 4 cmÀ1 . Gravimetric vapor sorption and interfacial energy (DVS) Moisture sorption profiles were obtained for the SD powders using a SGA-100 Sorption Analyzer (VTI Corporation, Hialeah, FL). The measurements were taken at 25 C (storage temperature) and 35 C (close to physiological temperature) using a sample size of 10–20 mg. All SD samples were exposed to an automated compu- terized sequence of increasing relative humidity (RH) in steps of 10% RH. The RH range was from 0% RH to 95% RH. The vapor sorption analysis was carried out to calculate the interfacial tension and water uptake capacity of all three powders as described in the literature27 . The adsorption of moisture onto multilayer surfaces is believed to follow the Brunauer–Emmett–Teller (BET) model. Similarly, we made the assumption that adsorption of moisture onto SD powder particles would follow the BET multilayer model. The BET constants (m0 and C) were determined from the initial lin- ear region of the moisture sorption adsorption isotherm (0.35% w/w). The interfacial tension at particle moisture interface was determined by calculating the binding energy (from BET constant C) and the average surface area of a water molecule (Appendix A). HPLC analytical method The quantitative analysis of model compounds used in the solubil- ity studies was performed by a reverse-phase gradient HPLC using an Agilent 1100 System (Agilent Technologies, Palo Alto, CA) equipped with a 4.6 Â 75 mm, 2.7 mm C18 column (Advanced Materials Technology, Wilmington, DE). The temperature of the col- umn was kept at 45 C, and the wavelength of detection was set to 210 nm. The mobile phase consisted of a binary gradient of Solvent A (acetonitrile) and Solvent B (HPLC grade water with 0.05% TFA). The linear gradient started at 5% A and increased to 95% A in 8.5 min, followed by a return to the starting condition within 1.6 min and equilibrated at the starting condition for 2 min. The injection volume was 10 mL. The flow rate was 1.0 mL/min, and the total run time was set for 13.10 min. This reverse-phase gradi- ent method provided a baseline resolution and excellent peak characteristics for the griseofulvin. Quantification of drug concen- trations was performed by analyzing the peak area at retention using Agilent ChemstationVR analytical software. Redispersion analysis The nanosuspensions have tendency to form loose or hard aggre- gates when spray-dried. Sometimes these aggregates are reversible and sometime they are not reversible. To evaluate this behavior, we carried out the redispersion study of SD NCs (with and without mannitol) in the DI water and in the simulated gastric media (pH 1.2). The study was performed in a conventional USP-type – II dis- solution apparatus at 100 rpm and 37 C temperature. Here, sam- ples were withdrawn at different time intervals and subjected to the particle size analysis using the zetasizer28 . In vitro dissolution rate determination A dissolution apparatus (Distek 2100C; Distek Inc., North Brunswick, NJ) equipped with a USP-type-II (paddle) was used to measure the dissolution rates of crystalline SD dispersions. The dissolution study was carried out by comparing the initial dissolution rates of MCs and NCs (with and without mannitol). Each dissolution study was carried out at 37 ± 0.5 C under a 100-rpm paddle speed in a USP- type II apparatus for 2 h. The dissolution was carried out under sink condition (sink condition factor 5.7Â), and under non-sink conditions (at 20%, 50%, and 90% saturation level of API in dissol- ution media) with a 50 mg loading of griseofulvin. A 10 mL of a dis- solution sample was withdrawn at intervals of 0, 2, 5, 7, 10, 20, 30, 45, 60, 90, and 120 min and replaced with the same volume of dis- solution media (pre-equilibrated at 37 ± 0.5 ). Samples were filtered through 0.1-mm (MCs) and 0.02-mm (NCs) anotop syringe filters (WhatmanTM ; GE Healthcare, Piscataway, NJ) and subjected to HPLC analysis. The first few milliliters of each sample were dis- carded to minimize losses because of adsorption. The discarded volume selected for each compound was determined by a separate filter validation experiment (data not shown). All studies were per- formed in triplicate29–35 . Results and discussion Morphological characterization The particle size of the suspensions before the spray dying was measured using DLS. The particle sizes of the SD powders were measured using the Sympatec GmbH System-Partikel-Technik (HELOS RODOS). The particle size and shape analysis data are presented in Table 1. The particle size of micronized SD powder upon drying is approximately 14 mm, while the particle size of both SD nanosuspension powders upon drying are around 6 mm. The particle size of nano powders are significantly lower than the particle size of the micronized powder, which is explainable due to difference in particle size before spray drying. There is no sig- nificant difference in particle size between nanosuspensions with and without mannitol. Morphological characterization by PLM and SEM (Figures 2 and 3) shows the particle size difference between MCs and NCs. Moreover, the NCs in both formulations became hollow upon particle size reduction while MCs appear cylindrical shaped. The SEM image of nanocrystals with mannitol (NCm) also shows the presence of mannitol on the surface of NCs. Initial observation from SEM images of the NCs with and without the mannitol showed formation of aggregates upon spray drying. Therefore, redispersion analysis was carried out for further characterization of the aggregation property (reversible or irreversible). The FLOWCHEM machine has the ability to capture 30 unique particle measurements. The morphological measure- ments for SD particles were calculated using Visual spread sheetVR particle analysis software. The NCm were found to be spherical in shape as compared to NCs without the mannitol and the MCs (Table 1). Solid-state characterization It is known that changes in the crystalline state can occur during both milling and spray drying. To ensure that the changes are due DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 3 Downloadedby[LIULibraries]at07:0905May2016
  • 5. to particle size reduction and that no amorphous griseofulvin was formed, MDSC, XRPD, and FT-IR were performed on the SD samples. Griseofulvin showed distinct crystalline peaks for 2h at 11 and 17 (Figure 4(a)) in all the SD powders. As shown by the XRPD pat- terns (Figure 4(a)), the reduction in the intensity and broadening of Figure 2. PLM images of (a) pure griseofulvin, (b) MC, (c) NCs, and (d) NCm. Figure 3. SEM images of (a) HPC-SL, (b) DOSS, (c) mannitol, (d) pure griseofulvin, (e) MC, (f) NCs, and (g) NCm. Table 1. Particle size and shape analysis. Formulation Avg. particle size before spray drying PDI Avg. particle size after SD (lm) Span(D90–D10)/D50 Circularity (Hu) Aggregation based on SEM/PLM MC 11 lm – 14.9 2.3 0.81 Yes NCs 210 nm 0.210 6.64 1.85 0.82 Yes Nanocrystal-mannitol 205 nm 0.192 6.42 1.82 0.95 Yes 4 D. A. SHAH ET AL. Downloadedby[LIULibraries]at07:0905May2016
  • 6. the peaks is observed for the nanoparticles. XPRD of SD powders showed a small hump in between 20 and 30 at 2h scale represents possible minor amorphous conversion. Figure 4(b) shows the MDSC thermograms for pure griseofulvin and the SD formulations. The thermograms were compared to confirm the crystallinity upon mill- ing and spray drying. The onset melting temperature of griseofulvin and mannitol were detected at 219.0 C and 168.4 C, respectively. After milling and spray drying, a reduction in the onset of the melt- ing temperature (2–4 C) and the heat of fusion was observed for both griseofulvin and mannitol. The melting peaks of griseofulvin shifted in the SD formulations (Figure 4(b)). The observed melting point reduction and peak shift have been reported to be in agree- ment with the particle size reduction. It can also be explained by the Gibbs–Thomson equation36 , which explains that, the melting temperature of a material is proportional to its cohesive energy. Since the atoms at the surface have reduced cohesive energy com- pared to bulk material due to physical milling and spray drying. Therefore, they require less energy to free from the solid phase (i.e. melting), which results in the reduction of melting point. Based on the MDSC and XRPD data, it can be concluded that the wet-media milling and spray drying might have affected the crystalline struc- ture of the model compound by partially (minor) converting it in to the amorphous form. For further clarification of the crystalline struc- ture, FT-IR spectroscopy was conducted on the SD samples (Data not shown). The SD formulation with mannitol showed intense peak at 3288À1 cm and 3388À1 cm (–OH stretching) suggesting the pres- ence of mannitol in the formulation. These transmissions were absent in the IR spectra of MCs and NCs without the mannitol. The IR spectra of all three SD formulations are same except the increase in the –OH and –CH stretching intensity due to the presence of mannitol in the SD formulation with mannitol. Interfacial energy and water uptake tendency The dynamic vapor sorption analysis was performed to evaluate the moisture uptake tendency of the SD powders. The water uptake tendency of NCm was found to be higher compared to both the MCs and NCs. For a detailed evaluation of this, we calculated the interfacial energy as described in Appendix A, and the results are presented in Table 2. For the colloidal dispersion systems, the inter- facial energy plays an important role. For different compounds, the solubility and, eventually, the dissolution rate increased as the sol- id–liquid interfacial tension decreases37 . The interfacial tension val- ues for the MC and NC do not have any significant difference at either temperature (both at 25 C and 35 C). The NCm showed less interfacial tension values compared to MCs and NCs without manni- tol at both temperatures. Here, mannitol has a characteristic prop- erty to act as a solubilizing and dispersing agent. Mannitol acts by forming capillaries in contact with aqueous solutions and generates rapid dispersion and dissolution38 . Based on these studies, we can conclude that the dissolution and redispersion behavior for NCm will be significantly higher due to the enhancement of the wetting property and a reduction in interfacial tension. Crisp39 and his col- leagues have also reported that the dissolution of NCs is more dependent on interfacial tension than the surface area. Redispersion analysis The nanosuspensions have a tendency to form loose or hard aggregates when SD. Nanoparticle aggregation was visually observed for SD powders in SEM images. Sometimes these aggre- gates are reversible and sometime they are not reversible. In order to assess their aggregation property, the redispersion analysis was carried out. The SD powder of NCm redispersed quickly during the redispersion study in water and in pH 1.2 (Figure 5(a) and (b)). The NCm quickly reached the initial particle size. On the other hand, the NCs without the mannitol were not able to achieve the initial particle size (200–300 nm) when spray-dried. The dispersion behav- ior of particles in the liquid phase is mainly affected by the inter- action between the particles and the dispersion media. Therefore, the presence of mannitol aids in wetting the particles quickly by forming reversible aggregates. In the case of the NCs without the Figure 4. Solid-state characterization and comparison of pure API, physical mixture, MCs, NCs, and NCm (a) XRPD, and (b) DSC. Table 2. Determination of BET constants and interfacial tension. Formulation Particle size (lm) C (binding constant) m0 (monolayer g/g solids) DE (J/mole) (sur- face binding energy) c (mN/m) (interfacial tension) 25 C 35 C 25 C 35 C 25 C 35 C 25 C 35 C MC 14.9 À20.58 À20.43 12.15 1.77 7492 7726 138.2 142.5 NCs 6.64 À17.00 À17.26 19.60 24.07 7681 7790 133 134 Nanocrystals-mannitol 6.42 À11.48 À11.24 32.26 21.19 6047 6195 111.6 114.3 DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 5 Downloadedby[LIULibraries]at07:0905May2016
  • 7. mannitol, they showed different wetting behavior in both water and pH 1.2, resulting in a significantly different redispersion profile. One possible reason might be the generation of nanocrystalline particles with a high adhesive hydrophobic surface, which has more of a tendency to form aggregates in the absence of the dis- persion agent (mannitol). Initial dissolution rate determination The dissolution studies were performed to discriminate the effect- iveness of the micro suspension, nanosuspension, and nanosuspen- sion with mannitol upon spray drying. The in-vitro dissolution profile of a formulation can serve as a predictive tool for in-vivo analysis40,41 . Here, the energy of the formulation system is reduced upon spray drying and with the rapid removal of water. For nano- particles, the rate limiting and more important step is the solvation step (surface kinetics) at the solid–liquid interface (the dissociation of drug molecules from the solid)39,42 . Therefore, the Noyes–Whitney model seems to be ineffective in generating the discrimination in the dissolution process of the NCs with a mean- ingful way. Our objective was to design a conventional in-vitro dissol- ution method, which can be easily applied to discriminate the dissolution profiles among the MCs, NCs, and NCm. Initially, the dissolution studies were carried out by maintaining the sink con- dition as described in the Method section (n ¼ 3). The sink con- dition dissolution profile did not show significant discrimination among MCs, NCs, or NCm (Figure 6(a)). Therefore, for efficient quantitative discrimination, the dissolution was carried out under non-sink conditions (n ¼ 3, Figure 6(a) and (b)). The non-sink dis- solution displayed discrimination in the dissolution profile by reducing the dissolution velocity. In the sink condition dissol- ution, the initial dissolution rate ratio of NCs and NCm is negli- gible. But, as the saturation level increased, the discrimination increased, and at the 90% saturation level, the initial dissolution rate ratio for NCs to MCs was 1.8, NCm to MCs was 6.7, and NCm to NCs was 4.9-fold compared to their dissolution ratios at initial phase in sink conditions (Table 3). The reported dissolution rate enhancement behavior can be attributed to the presence of mannitol during spray drying. This can be further explained by the improvement in the wetting property and a reduction of the interfacial tension (tension at solid–liquid interface during dissolution) for the SD particles of NCm. Also, the partial conver- sion of crystalline form to amorphous might play a contributing role in the dissolution behavior for the SD products. However, since the conversion of crystalline form to amorphous form is minor (probably less than 5%), the rate of crystallization of the amorphous form during dissolution expected to be very fast. Therefore, it will have negligible impact on the dissolution behavior. The current study justifies the use of non-sink condi- tion over sink condition for better quantitative discrimination of nanocrystalline and microcrystalline powder dissolution. Conclusion Much of the published work in the area of drying nanocrystalline suspensions has focused on the impact of the process parameters on the final product. This study shows the importance of particle Figure 5. Redispersion study of SD powders of NCs and NCm (a) in pH 1.2, and (b) water. Figure 6. In-vitro sink and In-vitro non-sink dissolution: comparing MCs, NCs, and NCm: (a) 0% and 20% saturation, and (b) 50% and 90% saturation levels. 6 D. A. SHAH ET AL. Downloadedby[LIULibraries]at07:0905May2016
  • 8. size reduction and the incorporation of mannitol as a dispersing agent during the spray drying of nanocrystalline suspension of poorly soluble compounds (BCS class II/IV). Upon spray drying, the fundamental properties (surface and in-vitro dissolution under non- sink condition) change drastically. The interfacial tension at the particle moisture interface and redispersion behavior and non-sink dissolution profile of griseofulvin–mannitol nanocrystalline suspen- sions show superior performance as compared to the micron/un- milled and nanocrystalline suspension formulations. The present study provides an in-depth understanding to a formulation scien- tist who can quantitate the change in the interfacial tension (at particle moisture interface) and in the in-vitro dissolution rate enhancement during initial phase. This provides a platform to apply a practical approach to screen and rank order the SD formu- lation with different initial particle size (before drying) with manni- tol as a dispersing agent. Disclosure statement The authors report no declaration of interests. References 1. Lipinski CA. Drug-like properties and the causes of poor solu- bility and poor permeability. J Pharmacol Toxicol Methods 2000;44:235–49. 2. Hywel D, Williams NLT, Charman SA, et al. Strategies to address low drug solubility in discovery and development. Pharmacol Rev 2013;65:315–499. 3. Shegokar R, Muller RH. Nanocrystals: industrially feasible multifunctional formulation technology for poorly soluble actives. Int J Pharm 2010;399:129–39. 4. Serajuddin ATM. Solid dispersion of poorly water-soluble drugs: early promises, subsequent problems, and recent breakthroughs. J Pharm Sci 1999;88:1058–66. 5. Dave RH, Shah DA, Patel PG. Development and evaluation of high loading oral dissolving film of aspirin and acetamino- phen. J Pharm Sci Pharmacol 2014;1:112–22. 6. Mou D, Chen H, Wan J, et al. Potent dried drug nanosuspen- sions for oral bioavailability enhancement of poorly soluble drugs with pH-dependent solubility. Int J Pharm 2011;413:237–44. 7. Merisko-Liversidge E, Liversidge GG, Cooper ER. Nanosizing: a formulation approach for poorly-water-soluble compounds. Eur J Pharm Sci 2003;18:113–20. 8. Murdande SB, Shah DA, Dave RH. Impact of nanosizing on solubility and dissolution rate of poorly soluble pharmaceuti- cals. J Pharm Sci 2015;104:2094–102. 9. Shah DA, Murdande SB, Dave RH. A review: pharmaceutical and pharmacokinetic aspect of nanocrystalline suspensions. J Pharm Sci 2016;105:10–24. 10. Kumar S, Jog R, Shen J, et al. Formulation and performance of danazol nano-crystalline suspensions and spray dried pow- ders. Pharm Res 2015;32:1694–703. 11. Song J, Wang Y, Song Y, et al. Development and character- isation of ursolic acid nanocrystals without stabilizer having improved dissolution rate and in vitro anticancer activity. AAPS PharmSciTech 2014;15:11–9. 12. M€uller RH, Peters K. Nanosuspensions for the formulation of poorly soluble drugs: I. Preparation by a size-reduction tech- nique. Int J Pharm 1998;160:229–37. 13. Rabinow BE. Nanosuspensions in drug delivery. Nat Rev Drug Discov 2004;3:785–96. 14. Cooper ER. Nanoparticles: a personal experience for formulat- ing poorly water soluble drugs. J Control Release 2010;141:300–2. 15. Van Eerdenbrugh B, Vermant J, Martens JA, et al. Solubility increases associated with crystalline drug nanoparticles: methodologies and significance. Mol Pharm 2010;7:1858–70. 16. Anhalt K, Geissler S, Harms M, et al. Development of a new method to assess nanocrystal dissolution based on light scat- tering. Pharm Res 2012;29:2887–901. 17. Ghosh I, Bose S, Vippagunta R, Harmon F. Nanosuspension for improving the bioavailability of a poorly soluble drug and screening of stabilizing agents to inhibit crystal growth. Int J Pharm 2011;409:260–8. 18. Chaubal MV, Popescu C. Conversion of nanosuspensions into dry powders by spray drying: a case study. Pharm Res 2008;25:2302–8. 19. Zuo B, Sun Y, Li H, et al. Preparation and in vitro/in vivo evalu- ation of fenofibrate nanocrystals. Int J Pharm 2013;455:267–75. 20. Isa L, Lucas F, Wepf R, Reimhult E. Measuring single-nanopar- ticle wetting properties by freeze-fracture shadow-casting cryo-scanning electron microscopy. Nat Commun 2011;2:438. doi:10.1038/ncomms1441. 21. Van Eerdenbrugh B, Froyen L, Van Humbeeck J, et al. Drying of crystalline drug nanosuspensions – the importance of sur- face hydrophobicity on dissolution behavior upon redisper- sion. Eur J Pharm Sci 2008;35:127–35. 22. Beck C, Sievens-Figueroa L, G€artner K, et al. Effects of stabil- izers on particle redispersion and dissolution from polymer strip films containing liquid antisolvent precipitated griseoful- vin particles. Powder Technol 2013;236:37–51. 23. Hu J, Ng WK, Dong Y, et al. Continuous and scalable process for water-redispersible nanoformulation of poorly aqueous soluble APIs by antisolvent precipitation and spray-drying. Int J Pharm 2011;404:198–204. 24. Niwa T, Danjo K. Design of self-dispersible dry nanosuspen- sion through wet milling and spray freeze-drying for poorly water-soluble drugs. Eur J Pharm Sci 2013;50:272–81. 25. Bojnanska E, Kalina M, Parizek L, et al. Determination of crit- ical parameters of drug substance influencing dissolution: a case study. BioMed Res Int 2014;2014. doi:http://dx.doi.org/ 10.1155/2014/929248. Table 3. Quantitation of initial dissolution rate ratios. Conditions Dissolved in 2 min (%) Initial dissolution rate ratios MC NC Nanocrystals-mannitol NCa /MCb NCmc /MC NCm/NC Sink 31 83 96 2.7 3.1 1.1 20% 11 33 37 3.0 3.3 1.1 50% 6 13 30 2.16 5.0 2.3 90% 4 7 28 1.75 7.0 4.0 a NC spray-dried, b micro SD powder, c nanocrystal with mannitol spray-dried. DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 7 Downloadedby[LIULibraries]at07:0905May2016
  • 9. 26. Sahoo NG, Kakran M, Shaal LA, et al. Preparation and charac- terization of quercetin nanocrystals. J Pharm Sci 2011; 100:2379–90. 27. Okoye PC. To systemically characterize magnesium stearate polymorphs and practically evaluate their effect on physico- chemical properties of naproxen (BCS Class II) and acet- aminophen (BCS Class III) as model drugs. Edited by Pharmaceutical Sciences. Brooklyn (NY): Long Island University;2013:190. 28. Prajapati H, Dalrymple DM, Serajuddin AT. In vitro dispersion test can serve as a predictive method for assessing perform- ance of lipid-based drug delivery systems. J Excipient Food Chem 2013;4:111–25. 29. Sun J, Wang F, Sui Y, et al. Effect of particle size on solubility, dissolution rate, and oral bioavailability: evaluation using coenzyme Q(10) as naked nanocrystals. Int J Nanomed 2012;7:5733–44. 30. Chidambaram N, Burgess DJ. A novel in vitro release method for submicron-sized dispersed systems. AAPS PharmSci 1999;1:32–40 31. Kocbek P, Baumgartner S, Kristl J. Preparation and evaluation of nanosuspensions for enhancing the dissolution of poorly soluble drugs. Int J Pharm 2006;312:179–86. 32. Jinno J, Kamada N, Miyake M, et al. Effect of particle size reduction on dissolution and oral absorption of a poorly water-soluble drug, cilostazol, in beagle dogs. J Control Release 2006;111:56–64. 33. Ma Q, Sun H, Che E, et al. Uniform nano-sized valsartan for dissolution and bioavailability enhancement: Influence of par- ticle size and crystalline state. Int J Pharm 2013;441:75–81. 34. Liversidge GG, Cundy KC. Particle size reduction for improve- ment of oral bioavailability of hydrophobic drugs: I. Absolute oral bioavailability of nanocrystalline danazol in beagle dogs. Int J Pharm 1995;125:91–7. 35. Korang-Yeboah M, Rahman Z, Shah D, et al. Impact of formu- lation and process variables on solid-state stability of theo- phylline in controlled release formulations. Int J Pharm 2016;499:20–8. 36. Wu W, Nancollas G. A new understanding of the relationship between solubility and particle size. J Solut Chem 1998; 27:521–31. 37. Kaur J, Aggarwal G, Singh G, Rana AC. Improvement of drug soluility using solid dispersion. Int J Pharm Pharm Sci 2012;4:47–53 38. Li X, Vogt FG, Hayes D, Mansour HM. Design, characteriza- tion, and aerosol dispersion performance modeling of advanced spray-dried microparticulate/nanoparticulate man- nitol powders for targeted pulmonary delivery as dry powder inhalers. J Aeros Med Pulm Drug Deliv 2014;27:81–93. 39. Crisp MT, Tucker CJ, Rogers TL, et al. Turbidimetric measure- ment and prediction of dissolution rates of poorly soluble drug nanocrystals. J Control Release 2007;117:351–9. 40. Heng D, Cutler DJ, Chan HK, et al. What is a suitable dissol- ution method for drug nanoparticles? Pharm Res 2008;25:1696–701. 41. Kataoka M, Itsubata S, Masaoka Y, et al. In vitro dissolution/ permeation system to predict the oral absorption of poorly water-soluble drugs: effect of food and dose strength on it. Biol Pharm Bull 2011;34:401–7. 42. Shekunov BY, Chattopadhyay P, Seitzinger J, Huff R. Nanoparticles of poorly water-soluble drugs prepared by supercritical fluid extraction of emulsions. Pharm Res 2006;23:196–204. Appendix A The adsorption of moisture onto multilayer surfaces is believed to follow the Brunauer–Emmett–Teller (BET) model. Similarly, we make the assumption that adsorption of moisture onto spray-dried pow- der particles follows the BET multilayer model. In this assignment, an attempt is made to relate surface energy from BET to interfacial tension based on moisture adsorption onto powder particles. Determination of BET parameters The BET equation is given by: m m0 ¼ CX ð1 À XÞ½1 þ ðC À 1ÞXŠ (A1) From thermodynamics, the ratio of partial pressure of water and that of pure substance is represented by: aw ¼ p p0 ¼ X (A2) We can now rearrange Equation (A1) to obtain the multilayer model of BET as shown by: m ¼ awm0C ð1 À awÞ½1 þ awðC À 1ÞŠ (A3) where m is the moisture content in g/g solids; aw, is water activity; m0, is the moisture content of monolayer in g/g solids; C ¼ e DE = RT , the BET model is typically applicable up to 0.5aw. The experimental application of the BET multilayer model involves obtaining some initial values of water activity, aw, with correspond- ing moisture content values, m. The analytical method involves lin- earization of Equation (A3) to obtain: ½1 þ awðc À 1ÞŠ m0c ¼ aw ð1 À awÞm (A4) Finally, aw ð1 À awÞm ¼ aw c À 1 m0c þ 1 m0c (A5) where a plot of aw ð1Àaw Þm versus aw gives a straight line with slope as cÀ1 m0c h i and the intercept as 1 m0c. The BET parameters, m0, and C were experimentally obtained as m0 ¼ 1 slopeþintercept and C ¼ 1 interceptÃm0 . Determination of the cross-sectional area of the water molecule Assume monolayer saturable sites with each molecule of water spreading across the spray-dried powder particle. From quantum thermodynamics, the spatial separation between two water mole- cules is given by: V N 1=3 18x10À6 m3 6x1023 1=3 ¼ ð30x10À30 m3 Þ1=3 ¼ 3x10À10 m (A6) 8 D. A. SHAH ET AL. Downloadedby[LIULibraries]at07:0905May2016
  • 10. 3x10À10 m à 3x10À10 m ¼ 9x10À20 m2 ¼ 0:9x10À19 m2 The cross-sectional area of a water molecule is then derived as: Note: The actual cross-sectional area of a water molecule: 1.06  10À19 m2 . Determination of interfacial tension (c) between particle and moisture The molecular relationship between solid–liquid interface may be described by the changes in surface energy (DE). The deposition of monolayer of moisture on the powder molecules creates an inter- facial tension that is expressed based on Young–Laplace expression: DE ¼ cA where DE is the surface or binding energy of the monolayer; c is the molecular interfacial tension; and A is the molecular cross sec- tional area. If we assume non-interactive particle–particle behavior as espoused by BET, then the molecular interfacial tension between powder and water molecules could be estimated from the binding or surface energy at the saturable sites as given by: c ¼ DE NAvogadro à 1 AH20 (A7) where DE is monolayer binding or surface energy; NAvogadro is the Avogadro’s number, (6:02x1023 molecules=mol); AH20 is the cross-sectional area of water molecule (1:06x10À19 m2 ). In conclusion, the BET model allowed the estimation of powder-moisture interfacial tension through the construct of surface energy of saturable monolayer. DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 9 Downloadedby[LIULibraries]at07:0905May2016