ArticlePDF Available

Preparation of delayed-release multiparticulate formulations of diclofenac sodium and evaluation of their dissolution characteristics using biorelevant dissolution methods

Authors:

Abstract and Figures

Diclofenac sodium was used as a model drug for preparation of delayed-release (DR) multiparticulates, which were further processed into solid oral dosage forms such as capsules and tablets. Multiple unit pellets systems (MUPS) were prepared from different types of starter pellets (inert cores) including microcrystalline cellulose pellets, sugar spheres, isomalt pellets and novel calcium phosphate-based pellets. The study results showed that the material of the inert cores affected both mechanical properties of the drug-loaded pellets and the dissolution characteristic of the model drug. Biorelevant dissolution method carried out with the help of a pHysio-grad device allowed thorough examination of the developed formulations in the environment mimicking pH conditions along gastrointestinal tract. This method revealed significant differences between the formulations and their sensitivity to variable hydrodynamic conditions.
Content may be subject to copyright.
Journal of Drug Delivery Science and Technology 60 (2020) 101986
Available online 18 August 2020
1773-2247/© 2020 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Research paper
Preparation of delayed-release multiparticulate formulations of diclofenac
sodium and evaluation of their dissolution characteristics using biorelevant
dissolution methods
Daniel Zakowiecki
a
,
*
, Maja Szczepanska
b
, Tobias Hess
a
, Krzysztof Cal
b
, Barbara Mikolaszek
b
,
Jadwiga Paszkowska
c
, Marcela Wiater
c
, Dagmara Hoc
c
, Grzegorz Garbacz
c
,
d
a
Chemische Fabrik Budenheim KG, Rheinstrasse 27, 55257, Budenheim, Germany
b
Department of Pharmaceutical Technology, Faculty of Pharmacy, Medical University of Gdansk, Al. Gen. J. Hallera 107, 80-416, Gdansk, Poland
c
Physiolution Polska Sp. Z o.o., Skarbowcow 81/7, 53-025, Wroclaw, Poland
d
Physiolution GmbH, Walther-Rathenau-Strasse 49a, 17489, Greifswald, Germany
ARTICLE INFO
Keywords:
Multiple unit pellets systems
Calcium phosphate-based pellets
Diclofenac sodium
Starter pellets
pHysio-grad
ABSTRACT
Diclofenac sodium was used as a model drug for preparation of delayed-release (DR) multiparticulates, which
were further processed into solid oral dosage forms such as capsules and tablets. Multiple unit pellets systems
(MUPS) were prepared from different types of starter pellets (inert cores) including microcrystalline cellulose
pellets, sugar spheres, isomalt pellets and novel calcium phosphate-based pellets. The study results showed that
the material of the inert cores affected both mechanical properties of the drug-loaded pellets and the dissolution
characteristic of the model drug. Biorelevant dissolution method carried out with the help of a pHysio-grad
device allowed thorough examination of the developed formulations in the environment mimicking pH condi-
tions along gastrointestinal tract. This method revealed signicant differences between the formulations and
their sensitivity to variable hydrodynamic conditions.
1. Introduction
Multiparticulate drug delivery systems (MDDS) are novel pharma-
ceutical dosage forms, which have been gaining increasing popularity in
recent years when compared to single unit dosage forms. These multiple
unit dosage forms (MUDF) are made of numerous independent subunits
(microparticles), where each of them is an autonomous reservoir of a
drug and releases the drug in a desirable way, independently of the other
subunits. Multiparticulates are especially suitable for preparation of
modied-release solid oral dosage forms (delayed- or sustained-release),
which offer such benets as less variable gastrointestinal transit or
reduced risk of dose dumping. At the present time, the most commonly
used multiparticulates are coated pellets which are formulated into oral
dosage forms either by lling them into hard gelatin capsules or com-
pacting into tablets (Multiple Unit Pellet System, MUPS) [14].
One of the well-established ways of preparation of multiparticulates
is drug layering of spherical starter pellets (also called inert cores or
beads) followed by coating these drug-loaded pellets with a thin
polymer-based lm. Drug layering of inert cores offers relatively easier
technology compared to extrusion and spheronisation process. More-
over, it enables the production of multiparticulates with a very uniform
particle size distribution as well as advantageous surface morphology [5,
6]. There is a wide range of various types of neutral starter pellets
commercially available on pharmaceutical market offering different
characteristics [7,8]. Sugar spheres (nonpareils), which were introduced
on the market and applied for the production of multiparticulates as
rst, are still the most popular. Sugar spheres are made of sucrose
(usually up to 92%) and corn starch [9]. They are soluble in aqueous
media and hygroscopic, which is often a challenge during coating [10].
Absorbed water can be problematic and impact stability of moisture
sensitive drugs [8]. That is why alternative products have been intro-
duced to the market and interest in their use is constantly growing. The
most commonly employed substitutes are cellulose pellets consisting of
100% microcrystalline cellulose (MCC). They are insoluble in water and
are characterized by high sphericity as well as good mechanical
strength. Due to these properties, the drug layering can be carried out
faster and with reduced formation of undesirable agglomerates, which
shortens the coating time and reduces cost of production. However,
* Corresponding author.
E-mail address: daniel.zakowiecki@budenheim.com (D. Zakowiecki).
Contents lists available at ScienceDirect
Journal of Drug Delivery Science and Technology
journal homepage: www.elsevier.com/locate/jddst
https://doi.org/10.1016/j.jddst.2020.101986
Received 2 April 2020; Received in revised form 31 July 2020; Accepted 31 July 2020
Journal of Drug Delivery Science and Technology 60 (2020) 101986
2
problems such as the absorption of certain drugs on the surface of cel-
lulose bers or swelling in contact with water, which can affect disso-
lution pattern have been reported [1113]. Another solution available
on the market are starter pellets consisted of polyalcohols such as iso-
malt, mannitol, xylitol. Similarly to sugar spheres, they are hygroscopic
and soluble in aqueous media, however, they are promoted due to their
low glycemic index and lack of cariogenic effect [5,14]. Within this
group, isomalt pellets offer especially interesting functionality of
creating internal osmotic pressure, which modulates dissolution pattern
of some drugs [15]. Recently, a new type of water-insoluble pellets
based on anhydrous dibasic calcium phosphate (DCPA) has been intro-
duced to the market. These pellets consist of 80% w/w of DCPA and 20%
w/w of MCC. Due to their elevated density of above 1000 g/l, very low
water content of less than 1%, and reduced hygroscopicity, they repre-
sent an attractive alternative to the other commercial products offered
so far. A detailed comparison of various functional properties of calcium
phosphate-based pellets with other commercially available inert cores
can be found elsewhere [16].
Inert cores are made of excipients commonly used in the pharma-
ceutical industry, which are neutral and should not exhibit any phar-
macological activity nor interact with the drug substance in a way,
which may adversely affect its stability and/or effectiveness. Never-
theless, characteristics of starter pellets may have a signicant impact on
the course of the production process as well as can inuence the rate of
drug dissolution not less than the properties of coating polymer. Thus,
the drug release pattern can be determined by the thickness of the
coating and its permeability, but also by the geometry of the pellets,
surface morphology or the osmotic pressure originating from the pellets
cores [5,8,15,1720].
As has been already mentioned, drug-loaded pellets can be lled into
hard gelatin capsule shells or compressed into tablets. Especially, the
latter technology of tabletting of microparticulates is very interesting
and allows combining the advantages of both tablets and pellets-lled
capsules in one dosage unit [2123]. In this case, the properties of
pellet cores together with the coating polymer will determine the
durability of the drug-loaded pellets and protect them from damages
during tabletting. It has been frequently reported that the size of pellets
is a very important factor to be considered during compaction. Nor-
mally, small pellets are employed in preparation of tablets as they are
less affected by the compression process. This is mainly due to the higher
mechanical strength of smaller beads relative to their size as well as
reduced contact force on each individual pellet resulting in signicantly
reduced degree of transformation [24,25].
In this work the suitability of different types of starter pellets in the
development of delayed-release multiparticulate formulations contain-
ing diclofenac sodium as a model drug was evaluated. Diclofenac so-
dium is a nonsteroidal anti-inammatory drug (NSAID) having both
analgesic and antipyretic activities. The drug is commonly used in the
treatment of rheumatic disorders. Diclofenac sodium is available in a
number of preparations of 25 mg, 50 mg or 75 mg strength [2628]. The
drug is a weak acid of BCS Class II and shows pH-dependent solubility in
physiological pH range which increases with the increase of pH value
[29,30]. Diclofenac sodium is often formulated as enteric-coated prep-
aration to avoid direct contact of the drug with the mucosa resulting in
local gastrointestinal toxicity. For this purpose pH-sensitive enteric
polymers are commonly used. These polymers should limit the release of
the drug in acidic conditions of stomach and allow rapid dissolution of
the drug in duodenum and distal parts of the gastrointestinal tract (GIT)
[31].
The development of drug products is a time-consuming process and
involves high costs. Thorough examination of drug formulations with
the help of reliable in vitro biorelevant techniques facilitates screening
of the candidates and makes this process more efcient. Biorelevant
dissolution methodologies allowing an adequate prediction of the in
vivo performance of drugs come here with help [32,33]. In the present
work a novel device, pHysio-grad, which enables biorelevant simulation
of the intestinal pH was employed [34]. The pHysio-grad can precisely
monitor and adjust the pH value of hydrogen carbonate buffer, which is
considered to be one of the most biorelevant medium for simulation of
intestinal conditions [35,36]. The carbonate buffer is a complex system
and its pH is an effect of dynamic interplay between all its constituents.
The thermodynamic instability leads to spontaneous increase of pH due
to the loss of carbon dioxide (CO
2
). This effect might be minimized by
either preventing CO
2
evaporation (by using appropriate sealing or
covering a dissolution medium with an organic layer) or by compen-
sating the loss of CO
2
with the help of an automated system. Moreover,
carbonate buffers give the opportunity for simulation of dynamic
intraluminal pH changes, however, continuous and dynamic adjustment
of pH value must be provided in order to use them in routine dissolution
testing.
The main objective of this work was to investigate the suitability of
different types of starter pellets in the development of delayed-release
multiparticulate formulations containing diclofenac sodium as a model
drug. The planned scope of research included the use of commercial
inert cores made of microcrystalline cellulose, sugar, isomalt as well as
novel calcium phosphate-based pellets in preparation of both hard
gelatin capsules and compressed tablets. The additional objective was to
examine and compare the effect of the core material on the in vitro drug
release of the model drug using compendial as well as biorelevant
dissolution methods.
2. Materials and methods
Diclofenac sodium (Amoli Organics, Mumbai, India). Film coating
systems: Vivacoat® FM-1M 000 (JRS Pharma, Rosenberg, Germany) and
Aquarius® Control ENA (Ashland, Covington, KY, USA). Inert cores:
calcium phosphate-based (DCPA) pellets - PharSQ® Spheres CM M
(Chemische Fabric Budenheim, Budenheim, Germany), microcrystalline
cellulose pellets - VIVAPUR® MCC Spheres 500 (JRS Pharma, Rosen-
berg, Germany), sugar spheres - pharm-a-spheresMESH 35-25 (Hanns
G. Werner GmbH, Tornesch, Germany), isomalt starter pellets - gale-
nIQ960 (BENEO-Palatinit GmbH, Mannheim, Germany). Microcrys-
talline cellulose VIVAPUR® 200 (JRS Pharma, Rosenberg, Germany).
Low-substituted hydroxypropyl cellulose (L-HPC) LH-11 (ShinEtsu,
Wiesbaden, Germany). Magnesium stearate (Peter Greven Fett-Chemi,
Venlo, The Netherlands). Transparent hard gelatin capsule shells, size
“0(Kapselwelt, Hude, Germany).
2.1. Preparation of drug-loaded DR pellets
The starter pellets used in this study were initially calibrated be-
tween two sieves, 500
μ
m and 710
μ
m, in order to obtain grains of
similar dimensions and to avoid the effect of different particle sizes on
the coating process as well as on analyses results. Sieved pellets were
drug-layered with diclofenac sodium in a ProCepT 4M8-Trix Fluid-bed
system (FBS) equipped with a Wurster column (ProCepT nv, Zelzate,
Belgium). Around 100 g of starter pellets were rst coated with a
aqueous suspension containing 5% w/w of the drug substance and 5%
w/w of Vivacoat® system up to about 20% of weight gain. Subsequently,
without breaking the process, the pellets were sprayed with puried
water (intermediate coating) and nally coated with 20% w/w aqueous
suspension of Aquarius® Control ENA (enteric-coating) until around
10% of weight gain was reached. The use of an intermediate coating step
allowed maintaining the continuity of the entire process and adjusting
the process parameters before the following coating phase. Moreover, it
reduced interactions between the two layers and the accumulation of
static charges. During all phases the machine settings were maintained
at the following values: air speed of around 0.21 m
3
/min, inlet air
temperature of around 63 C, product temperature of around 40 C,
nozzle pressure of 1 bar.
D. Zakowiecki et al.
Journal of Drug Delivery Science and Technology 60 (2020) 101986
3
2.2. Preparation of multiparticulate dosage forms
Enteric-coated pellets containing the model drug were used to
formulate two types of solid oral dosage form - hard gelatin capsules and
compressed tablets. Developed MUPS formulations consisted of around:
50.0% w/w of the diclofenac sodium pellets (equivalent to 25 mg of
diclofenac sodium), 44.5% w/w of MCC type 200, 5.0% w/w of L-HPC
and 0.5% w/w of magnesium stearate. Hard gelatin capsules were pre-
pared by manual lling of the diclofenac sodium pellets into capsule
shells of 0size. Tablets were compressed on a Korsch EK0 eccentric
tablet press (Korsch, Berlin, Germany) using concave punches (R 25
mm) of 12 mm diameter under a compaction forces of 1820 kN.
2.3. Scanning electron microscopy
To visualize surface properties of the drug-loaded DR pellets alone,
as well as compressed into tablets, a scanning electron microscope
Phenom Pro (Phenom World Thermo Fisher, Eindhoven, Netherlands)
was employed. Standard sample holder and a carbon tape were used to
x a sample and acceleration voltage of 510 kV was applied to record
images at a magnication of 300x.
2.4. Mechanical strength
The mechanical strength of starter pellets as well as drug-loaded DR
pellets was assessed with a texture analyzer equipped with a 6 mm
diameter cylindrical stainless steel probe in compression mode (TA.XT
Plus, Godalming, Surrey, England). The probe was moved vertically
downwards at a speed of 2.0 mm/s until the triggering force of 0.1 N was
detected and further with a speed of 0.05 mm/s until a pellet was
crushed. The maximum force at the point of fracture and the distance to
break were recorded. All measurements (for each type of sphere n =12)
were performed at room temperature.
2.5. Dissolution test (modied compendial method)
According to The United States Pharmacopeia monograph for
diclofenac sodium delayed-release tablets, dissolution test is carried out
in two steps, between which test samples must be transferred from the
one medium to the other. In the case of enteric-coated tablets a paddle
apparatus is applicable because the transfer of such tablets is not a major
problem. However, in the case of microparticulates the use of a basket
apparatus is more convenient. In this study both hard gelatin capsules
and tablets containing enteric-coated diclofenac sodium pellets were
placed in a basket apparatus (USP apparatus 1) PTWS 820D (Phar-
maTest AG, Hainburg, Germany) and the dissolution test was carried out
according to the monograph for Diclofenac Sodium Delayed-Release
Tablets given in USP42/NF37 using a rotational speed of 100 rpm.
During the rst phase the capsules were incubated in 0.1 M hydrochloric
acid for 2 h. In acidic stage, according to both USP/NF and Ph.Eur.
gastro-resistant pellets should not release more than 10% of labeled
content of a drug. In the subsequent buffer stage, release of the drug
should be rapid and reach minimum 80% of the labeled amount of
diclofenac sodium within 45 min. The amount of the dissolved drug was
determined using a T70 UV/VIS Split-Beam Spectrophotometer (Phar-
maTest AG, Hainburg, Germany) at the detection wavelength of 276 nm
using ow-through quartz cuvettes with 1 cm path length.
2.6. Biorelevant dissolution in hydrogen carbonate buffer using a pHysio-
grad device
The pHysio-grad device used in this study was composed of a
microcomputer, which controlled independently each measurement
channel and a valve island. pH probes assured constant pH measurement
in each vessel and triggered adjustment of pH when necessary. The
adjustment of pH values was obtained by addition of a titrant, which was
CO
2
and compressed air in this case (Fig. 1).
Dissolution was carried out in a paddle apparatus (USP apparatus 2)
at a 50 rpm paddle rotational speed. Dissolution test was composed of
two stages. In the rst stage tablets or pellets (taken out from capsule
shells) were placed in a vessel lled with 250 mL of 0.01 M HCl for 30
min. After this time 750 mL of Hanks buffer concentrate was added and
the test continued for 60 min. After addition of buffer, the pH of
hydrogen carbonate medium was controlled by pHysio-grad device and
set at 6.8. The amount of the dissolved drug was determined using an
Agilent 8453 UV/VIS Spectrophotometer (Agilent Inc. Santa Clara, CA,
USA) at the detection wavelength of 276 nm using quartz cuvettes with
1 cm path length.
3. Results
3.1. Mechanical strength
Mechanical strength of starter pellets and drug-loaded DR pellets was
assessed with a texture analyzer. The maximum force at the point of
fracture (hardness) and the distance to break vs. pellets breaking
strength [N] are shown in Figs. 2 and 3. The highest value of hardness
was recorded for MCC spheres, and the lowest for sugar-based inert
cores. Generally, it was observed that for all types of starter pellets the
coating process signicantly improved the mechanical strength of the
particles. The increase in robustness was the most pronounced in case of
water-insoluble pellets (MCC and DCPA). It can be noted that for iso-
malt-, sugar- and MCC-based pellets values of the standard deviation are
relatively high which could result from their not quite spherical shape.
Drug-loaded enteric-coated pellets based on isomalt and sugar exhibited
very similar response to the compression. They deformed relatively fast
when exposed to external force until reaching the point of fracture as
shown in Fig. 3. Deformation of DCPA-based pellets was signicantly
slower. It can be noted that the maximum force at the breaking point is
at similar level as for isomalt pellets, but the distance to reach fracture
point was signicantly extended. This can be the effect of the specic
composition of DCPA-based inert cores, which combine the features of
brittle calcium hydrogen phosphate and a ductile material (microcrys-
talline cellulose).
3.2. Scanning electron microscopy
Surface structure and cross-sections of the drug-loaded DR pellets
before and after compression into tablets were studied with a scanning
electron microscope. Fig. 4 shows SEM micrographs of drug-loaded
diclofenac sodium pellets before compression (left column), com-
pressed pellets inside tablets (middle column) and pellets extracted from
the surface of tablets (right column). SEM images of cross-sections show
Fig. 1. Schematic diagram of the pHysio-grad device.
D. Zakowiecki et al.
Journal of Drug Delivery Science and Technology 60 (2020) 101986
4
that the shape of sugar- and DCPA-based drug-loaded pellets is spher-
ical, whilst isomalt and MCC show more ellipse-like character. In all
cases a clear distinction between a core and a coating layer can be
spotted with no visible deformation of the coating layer. The coating
layer is uniform through the whole cross-sections with layer thickness of
approximately 30
μ
m. Although the irregular shape of the some types of
examined inert cores could have caused a variation in the coating
thickness, no such observations were made. It can be seen that even after
compression the original shape of the pellets located inside the tablets
generally remained unchanged, without visible deformations or dam-
ages of the coating layer. However, it should be noted here that many
pellets positioned on the very surface of the tablets, which were in direct
contact with the punches during compaction, displayed cracks in the
polymer layer and partial shape deformation. The most pronounced
cracks are visible for pellets made of sugar, isomalt and MCC. Especially
sugar-based pellets were heavily deformed, which led to partial expo-
sure of their cores.
3.3. Dissolution test (modied compendial method)
Four types of commercially available inert cores (DCPA pellets, MCC
spheres, sugar spheres and isomalt pellets) were used to prepare
delayed-release multiparticulate formulations (hard gelatin capsules
and tablets) of diclofenac sodium at a dose of 25 mg. All of them were
analyzed using modied compendial methods for diclofenac sodium
delayed-release tablets (USP42/NF37). Comparison of the amount of the
drug released in the acidic medium is given in Fig. 5. During this phase
both hard gelatin capsules and tablets disintegrated completely and
Fig. 2. Comparison of breaking strength (hardness) of inert cores made of DCPA, MCC, sugar and isomalt with corresponding drug-loaded DR pellets (given are
means of min. 10 repetitions, SD is indicated by the error bars).
Fig. 3. The distance to break and the maximum force at the point of fracture of drug-loaded enteric-coated pellets based on DCPA, MCC, sugar and isomalt (given are
examples of single deformation proles).
D. Zakowiecki et al.
Journal of Drug Delivery Science and Technology 60 (2020) 101986
5
water-insoluble ingredients formed a cone at the bottom of dissolution
vessels. In the baskets remained practically only pellets with very small
residue of excipients. It was observed that the water-soluble inert cores
(made of sugar and isomalt) tended to oat in the upper part of baskets,
whereas the insoluble ones (DCPA and MCC) remained mostly at the
bottom of baskets. All examined formulations demonstrated their
gastro-resistance and released much less than 10% of the labeled
amount of diclofenac sodium within 2 h of the test. However, in case of
preparations containing water-soluble starter pellets higher dissolution
of the drug substance can be detected. It can also be noticed that tablets
containing inert cores with MCC, sugar and isomalt released less amount
of the drug than analogous capsules. The opposite situation occurred in
case of phosphate pellets, where the release of diclofenac sodium from
tablets is slightly higher than from capsules.
Fig. 4. SEM micrographs of drug-loaded diclofenac sodium DR pellets (magnication of 300x).
Fig. 5. The release of diclofenac sodium from gastro-resistant diclofenac sodium preparations in acid phase (after 2 h incubation in 0.1 M HCl) (given are mans of n
=6, SD is indicated by the error bars).
D. Zakowiecki et al.
Journal of Drug Delivery Science and Technology 60 (2020) 101986
6
In the buffer stage all tested formulations demonstrated rapid
dissolution and released the whole amount of the drug within required
45 min (Fig. 6). It can be noted that multiparticulates containing water-
soluble substances showed identical, measurably faster dissolution rate
in comparison to insoluble MCC and DCPA cores. In the case of phos-
phate pellets, the release of diclofenac sodium from tablets is slightly
slower than from hard gelatin capsules, while in the other examined
formulations these differences are not so visible. After completing this
phase in the baskets containing DCPA and MCC microparticulates still
the whole quite spherical cores could be observed while for water-
soluble sugar and isomalt beads the baskets were practically empty. In
the case of MCC spheres, a large amount of retained red dye (which is a
component of the enteric-coating) can be seen. (Fig. 7).
3.4. Biorelevant dissolution in hydrogen carbonate buffer using a pHysio-
grad device
Biorelevant dissolution test has shown the high discriminatory
power and revealed signicant differences between the tested formu-
lations in terms of the impact of the dosage form as well as the core
material on the dissolution rate. Moreover, the use of a paddle apparatus
enabled visual observation of the tested samples directly in dissolution
vessels.
The results presented in Fig. 8 show that drug-loaded pellets con-
taining sugar and isomalt, both alone (without capsule shells) and
compressed into tablets, released diclofenac sodium faster than corre-
sponding MCC and DPCA cores. In addition, the release of the model
drug from these tablets and free pellets was virtually identical and the
dosage form had no effect on its dissolution rate. These ndings are in
line with the results obtained in the dissolution test based on the com-
pendial method (Fig. 6). A different situation can be observed in the case
of pellets containing water-insoluble MCC and DCPA. Here, the differ-
ences between the two dosage forms are clearly visible. The dissolution
rate from pellets is much slower than from corresponding tablets. This
difference is noticeably larger in the case of DCPA cores.
Examination of free pellets allowed observation of their different
dissolution behavior which might be related to the core material. Sugar-
and isomalt-based pellets were mobile and tended to oat in the entire
volume of dissolution medium. Dosing of a gaseous titrants in the second
stage of the test additionally stimulated the movement of pellets in the
vessel which could promote dissolution of diclofenac sodium. On the
contrary, MCC- and DCPA-based pellets exhibited more stationary
behavior. Especially pellets containing DCPA, due to their elevated
density, were lying still at the bottom of the vessels even during addition
of gaseous titrants. The limited movement resulted in much lower
dissolution rate, particularly in comparison with sugar and isomalt
cores. DCPA-based pellets did not release the whole of the labeled
amount of diclofenac sodium within 60 min of the test. Additional
intensive mixing was needed to release all the substance (results not
included in the diagram).
Signicant differences in dissolution pattern between tablets and
free pellets could be observed, especially at the very beginning of the
second stage of the dissolution test. First samples were withdrawn in 1
min after addition of Hanks buffer (Fig. 9). All tested tablets have
released more than 20% of label amount of diclofenac sodium within
this time, which could be caused by the presence of excipients that
promoted movement of freed pellets and increased their exposure to the
dissolution medium. For comparison, at the same time the free pellets
released a much smaller amount of the drug substance (about 1213%
for sugar and isomalt pellets, 11% for MCC cores and only 4% for DCPA
cores). In case of pellets based on water-insoluble material, contrary to
tablet formulations, free pellets showed less tendency to move and were
lying at the bottom of the dissolution vessels. It could be also observed
that dosing of the gaseous titrant had a greater effect on movement of
isomalt and sugar cores and facilitated their oating in the entire volume
of the dissolution medium.
4. Discussion
The aim of this work was to compare the suitability of different types
of starter pellets in the development of delayed-release multiparticulate
formulations containing the model drug - diclofenac sodium. Four types
of commercial inert cores of pharmaceutical grade were selected for the
research. Two of them were soluble in aqueous media sugar spheres
and isomalt pellets. Other two, microcrystalline cellulose spheres and
calcium phosphate-based pellets, were insoluble in water. Starter pellets
available on the market differ signicantly in size which can signi-
cantly impact properties of multiparticulate formulations including their
dissolution characteristics [3739]. In order to eliminate impact of
particle diameters on results of analyses the cores used in this study were
normalized and only the 500710
μ
m fraction was utilized. A chemical
nature of the core material determined not only the solubility but also
Fig. 6. Dissolution rate of diclofenac sodium from various multiparticulates in the buffer stage (given are mans of n =6, SD is indicated by the error bars).
D. Zakowiecki et al.
Journal of Drug Delivery Science and Technology 60 (2020) 101986
7
the functional properties of starter pellets that were inuencing the
process of drug layering of the cores. For instance, different tendencies
to accumulate electrostatic charges have been observed. Fig. 10 shows
pictures of FBS chamber taken about 5 min after the start of the process.
In case of MCC, sugar and isomalt cores, high adherence of the particles
to the glass walls of the granulator, caused by static electricity, is clearly
visible. Fluidization of DCPA beads is smooth and undisturbed. More-
over, in the case of water-soluble inert cores, especially made of isomalt,
initial extensive formation of dust has been noticed. This observation
corresponds to the data published earlier and showing higher friability
of these inert cores [16].
Starter pellets differed in terms of hardness expressed here as
breaking strength (see Fig. 2) The most durable were MCC spheres, the
lowest mechanical strength was observed for sugar- and DCPA-based
inert cores. The coating of inert cores in this case with a water sus-
pension of hydroxypropyl methylcellulose (HPMC) mixed with the equal
amount of diclofenac sodium signicantly improved mechanical
strength of the pellets. The greatest improvement was observed for
DCPA pellets for which hardness value increased by over 2.25 times. In
the course of research, no clear relationship between pellets hardness
and resistance to crush during tabletting could be observed. Considering
that the coating thickness for all pellets was identical (around 30
μ
m),
the differences in their robustness had to be due to the properties of the
core material. Drug-loaded enteric-coated sugar spheres with the lowest
value of breaking strength were affected the most during compression
(see Fig. 4). Nevertheless, when comparing coated pellets of similar
hardness, i.e. based on isomalt and DCPA, one can notice much less
compression damages of these latter. This difference is probably due to
Fig. 7. Residues of diclofenac sodium DR pellets (DCPA, MCC, sugar, isomalt) at the bottom of basket after completing the buffer stage.
Fig. 8. Dissolution rate of diclofenac sodium from various multiparticulates in the hydrogen carbonate buffer pH 6.8 (biorelevant test condition) (given are mans of
n =3 SD is indicated by the error bars).
D. Zakowiecki et al.
Journal of Drug Delivery Science and Technology 60 (2020) 101986
8
the longer time needed to reach the fracture point for the phosphate-
based pellets, as shown in Fig. 3. Furthermore, the inert cores made of
ductile MCC with the highest value of breaking strength were damaged
even more than previous two types. It should be mentioned here that the
pellets of the sizes used in this study (500710
μ
m) are normally not
employed for preparation of MUPS tablet. However, a similar approach
has been successfully applied for research purposes and reported by
Hiew et al. [40] In this case, however, such particle dimensions allowed
precise observation of the coating layer and its deformation or damages
arose during compression into tablets. It is also important that the
damages were limited to the pellets located on the very surface of the
tablets, which had direct contact with steel parts of tableting tooling. It
can therefore be assumed that many of the observed cracks were caused
mechanically and the use of larger amounts of cushioning excipients
could signicantly reduce the damages.
Diclofenac sodium is known to cause adverse GI side effect after oral
administration [29]. Thus its preparations are commonly enteric-coated
in order to reduce gastric exposure. Enteric-coating should limit release
of the active substance in the stomach as much as possible and then
allow quick dissolution in the duodenum and distal GIT. In the present
studies, the quality of the coating layer has been evaluated after pro-
longed storage under acidic conditions, reecting those prevailing in the
stomach, as it is recommended in Ph.Eur. (chapter 2.9.3.) or USP/NF
(chapter 711). After 2 h of maceration in 0.1 M HCl, it was found that all
tested microparticulates met the compendial requirements and did not
release more than 10% of the labeled content of the active substance (see
Fig. 5) proving their gastro-resistance. It can be noted, however, that the
drug-loaded pellets based on water-insoluble cores released less diclo-
fenac sodium than water-soluble ones. When comparing MUPS tablets
and capsules, one can also observe slight differences in the amount of the
drug substance dissolved. For tablets containing sugar, isomalt or MCC
cores, the release of diclofenac sodium under acidic conditions was
lower than from corresponding capsules. The smallest change was
observed in the case of microparticulates based on MCC. This observa-
tion is unexpected as the analysis of SEM micrographs (see Fig. 4)
revealed damages to the polymer layer of many pellets after compres-
sion. Probably during prolonged maceration in aqueous solution of hy-
drochloric acid, the polymer coating was hydrated and formed hydrogel
Fig. 9. The release of diclofenac sodium after 30 min incubation in 0.01 M HCl and 1 min after addition of hydrogen carbonate buffer (given are mans of n =3 SD is
indicated by the error bars).
Fig. 10. Chamber of uid bed system after around 5 min of the coating process containing 1) DCPA pellets, 2) MCC spheres, 3) sugar spheres, 4) isomalt pellet.
D. Zakowiecki et al.
Journal of Drug Delivery Science and Technology 60 (2020) 101986
9
which sealed microparticulates interior. Interestingly, however, this
phenomenon has a larger magnitude in the case of pellets compressed
into tablets which might result from the very close contact of
drug-loaded pellets surface with excipients caused by compaction
forces. DCPA-based DR pellets behaved differently and after compres-
sion into tablets released more active substance. This could be explained
by a less effective sealing of the enteric-coating during maceration. It
should be noted that the difference between tablets and capsules
formulation was not big and the amount of dissolved diclofenac sodium
in this case was at the same level as for MCC cores and furthermore,
much lower than for both sugar- and isomalt-based multiparticulates.
The assumption about the sealing of the coating layer is to a certain
degree justiable, taking into account the results shown in Fig. 9 where
four times shorter maceration in much smaller volume of diluted acid
did not cause such sealing effect and allowed much higher release of
active substance from tablets when compared to the corresponding free
pellets (taken out from capsule shells). The highest difference was
observed for DCPA-based microparticulates, nevertheless, it should be
noted that the release of diclofenac sodium from the tablets containing
calcium phosphate cores is lower that from other tested MUPS tablets.
For formulation containing MCC- and isomalt-based DR pellets, big error
values can be spotted. In the rst case, this may be due to the plastic
nature of the cores and their signicant deformation during compression
resulting in a different coating thickness. For isomalt-based drug-loaded
pellets this phenomenon is related to the high solubility of the core
material in water. From the very beginning of the dissolution test, both
uncompressed and compressed multiparticulates were partly oating on
the surface of the dissolution medium, where parts of the pellets were
not immersed in the medium.
Comparing results of the dissolution carried out in buffers of neutral
pH using modied compendial method in a basket apparatus (see Fig. 6)
and the biorelevant method in a paddle apparatus (see Fig. 8), a very
similar relationship between the drug release rate and the chemical
nature of inert cores was found: dissolution rate from water-soluble
sugar- and isomalt-based multiparticulates was much faster than from
insoluble ones. Furthermore, in the case of drug-loaded pellets based on
water-soluble inert cores the release of diclofenac sodium from both
tablets and hard gelatin capsules was insensitive to differences in hy-
drodynamic conditions generated in a paddle or a basket apparatus,
showing similarly fast dissolution rate, normally exceeding 85% of the
amount of the drug substance within rst 15 min of the test. The
dissolution rate from tablets containing MCC- and DCPA-based multi-
particulates was slightly slower and their release proles had a similar
shape. In the case of uncompressed DR pellets lled in capsules, a very
high susceptibility of the drug release to the hydrodynamic conditions in
an apparatus was found. Comparing the results obtained using the
basket and paddle methods, one can observe much slower dissolution
rate for the latter method. This is due to the fact that the water-insoluble
pellets cores, especially DCPA-based ones, had a higher density and
tended to lay on the bottom of dissolution vessel directly under the
paddles, where mixing intensity is normally the lowest. Interestingly
though, under these conditions the release prole observed for calcium
phosphate pellets was very close to 0-order kinetics.
5. Conclusions
In the course of this study, delayed-release multiparticulate formu-
lations of diclofenac sodium in the form of tablets and hard gelatin
capsules were developed. As a basis for the preparation of the multi-
particulates, the starter pellets containing sucrose, isomalt, microcrys-
talline cellulose and anhydrous dibasic calcium phosphate were used. It
has been shown that the core material signicantly inuenced the
coating process, the properties of drug-loaded pellets as well as their
dissolution behavior. For diclofenac sodium, it seems more advanta-
geous to use starter pellets based on water-insoluble substances such as
MCC or DCPA, which showed the lowest degree of the drug release in the
acid phase. This may result in lower concentration of diclofenac sodium
in the gastric lumen and reduce stomach irritation.
The tableting of drug-loaded enteric-coated pellets revealed slightly
different resistance to compression and susceptibility to mechanical
damages of investigated inner cores. It can be assumed, the use of starter
pellets of smaller size would result in obtaining more rugged multi-
particulates, more suitable for formulation into tablets as reported
elsewhere [37,39,41].
The dissolution rate of diclofenac sodium from developed formula-
tions in neutral conditions mimicking these in the distal GIT was clearly
related to the solubility of the core material in water very rapid for
soluble cores made of isomalt and sugar, slower for insoluble ones (MCC-
and DCPA-based).
References
[1] N. Al-Hashimi, N. Begg, R.G. Alany, H. Hassanin, A. Elshaer, Oral modied release
multiple-unit particulate systems: compressed pellets, microparticles and
nanoparticles, Pharmaceutics (2018), https://doi.org/10.3390/
pharmaceutics10040176.
[2] I. Ghebre-Selassie, Pellets: a general overview I, in: Pharmaceutical Pelletization
Technology, Marcel Dekker, 1989.
[3] S. Muley, T. Nandgude, S. Poddar, Extrusionspheronization a promising
pelletization technique: in-depth review, Asian J. Pharm. Sci. (2016), https://doi.
org/10.1016/j.ajps.2016.08.001.
[4] M. Zaman, S. Saeed-Ul-Hassan, R.M. Sarfraz, N. Batool, M.J. Qureshi, M.A. Akram,
S. Munir, Z. Danish, Pellets and pelletization: emerging trends in the pharma
industry, Acta Pol. Pharm. - Drug Res. 73 (3) (2016) 14151425.
[5] O. Luhn, N. K´
allai, Z.K. Nagy, K. Kov´
acs, B. Fritzsching, I. Klebovich, I. Antal,
Dissolution prole of novel composite pellet cores based on different ratios of
microcrystalline cellulose and isomalt, J. Pharm. Sci. (2012), https://doi.org/
10.1002/jps.23186.
[6] R. Deb, A.B. Ahmed, Pellets and pelletization techniques: a critical review, Int. Res.
J. Pharm. (2016), https://doi.org/10.7897/2230-8407.04414.
[7] P. Tschopp, Starting pellets: materials, manufacturing methods, and applications,
Tablets & Capsules Magazine 13 (5) (2015) 1013.
[8] R. Sidwell, J. Hansell, M. Rane, A.R. Rajabi-Siahboomi, Characterization of Inert
Cores for Multiparticulate Dosage Forms, 2017, https://doi.org/10.1007/978-1-
4939-7012-4_2.
[9] D. Werner, Sugar spheres: a versatile excipient for oral pellet medications with
modied release kinetics, Pharmaceut. Technol. Eur. 18 (4) (2006) 3541.
[10] E. Gryczov´
a, M. Rabiˇ
skov´
a, D. Vetchý, K. Krejˇ
cov´
a, Pellet starters in layering
technique using concentrated drug solution, Drug Dev. Ind. Pharm. (2008),
https://doi.org/10.1080/03639040802130046.
[11] S.S. Al-Nimry, S.M. Assaf, I.M. Jalal, N.M. Najib, Adsorption of ketotifen onto some
pharmaceutical excipients, Int. J. Pharm. (1997), https://doi.org/10.1016/S0378-
5173(96)04857-0.
[12] A.S. Narang, D. Desai, S. Badawy, Impact of excipient interactions on solid dosage
form stability, Pharm. Res. (N. Y.) 29 (2012) 26602683, https://doi.org/10.1007/
s11095-012-0782-9.
[13] S.L. Rivera, S. Ghodbane, In vitro adsorption-desorption of famotidine on
microcrystalline cellulose, Int. J. Pharm. (1994), https://doi.org/10.1016/0378-
5173(94)90413-8.
[14] G. Livesey, Health potential of polyols as sugar replacers, with emphasis on low
glycaemic properties, Nutr. Res. Rev. (2003), https://doi.org/10.1079/nrr200371.
[15] N. K´
allai, O. Luhn, J. Dred´
an, K. Kov´
acs, M. Lengyel, I. Antal, Evaluation of drug
release from coated pellets based on isomalt, sugar, and microcrystalline cellulose
inert cores, AAPS PharmSciTech (2010), https://doi.org/10.1208/s12249-010-
9396-x.
[16] D. Zakowiecki, M. Lachmann, V. Schaum, T. Hess, Novel starter pellets based on
dibasic calcium phosphate anhydrous: properties and application, Express Pharma
15 (1) (2019) 3436.
[17] S. Abdul, A.V. Chandewar, S.B. Jaiswal, A exible technology for modied-release
drugs: multiple-unit pellet system (MUPS), J. Contr. Release (2010), https://doi.
org/10.1016/j.jconrel.2010.05.014.
[18] A. Dashevsky, K. Kolter, R. Bodmeier, Compression of pellets coated with various
aqueous polymer dispersions, Int. J. Pharm. (2004), https://doi.org/10.1016/j.
ijpharm.2004.03.019.
[19] A.B. Bashaiwoldu, F. Podczeck, J.M. Newton, The application of non-contact laser
prolometry to the determination of permanent structural change induced by
compaction of pellets: II. Pellets dried by different techniques, Eur. J. Pharmaceut.
Sci. (2004), https://doi.org/10.1016/j.ejps.2004.02.005.
[20] C. Seiler, Oral controlled release technology and development strategy, in:
Pharmaceutical Formulation: the Science and Technology of Dosage Forms,
Croydon, Cambridge RSC, 2018, https://doi.org/10.1039/9781782620402.
[21] F. Zeeshan, K.K. Peh, Y.T.F. Tan, Exploring the potential of a highly compressible
microcrystalline cellulose as novel tabletting excipient in the compaction of
extended-release coated pellets containing an extremely water-soluble model drug,
AAPS PharmSciTech (2009), https://doi.org/10.1208/s12249-009-9278-2.
D. Zakowiecki et al.
Journal of Drug Delivery Science and Technology 60 (2020) 101986
10
[22] V.S.N. Murthy Dwibhashyam, J. Ratna, Key formulation variables in tableting of
coated pellets, Indian J. Pharmaceut. Sci. (2008), https://doi.org/10.4103/0250-
474X.45391.
[23] T. Chen, J. Li, T. Chen, C.C. Sun, Y. Zheng, Tablets of multi-unit pellet system for
controlled drug delivery, J. Contr. Release (2017), https://doi.org/10.1016/j.
jconrel.2017.07.043.
[24] J.L. Haslam, A.E. Forbes, G.S. Rork, T.L. Pipkin, D.A. Slade, D. Khossravi, Tableting
of controlled release multiparticulates, the effect of millisphere size and protective
overcoating, Int. J. Pharm. (1998), https://doi.org/10.1016/S0378-5173(98)
00240-3.
[25] B. Johansson, F. Nicklasson, G. Alderborn, Effect of pellet size on degree of
deformation and densication during compression and on compactability of
microcrystalline cellulose pellets, Int. J. Pharm. (1998), https://doi.org/10.1016/
S0378-5173(97)00355-4.
[26] R.E. Small, Diclofenac sodium, Clin. Pharm. (1989), https://doi.org/10.2165/
00003495-198835030-00004.
[27] P.A. Todd, E.M. Sorkin, Diclofenac sodium: a reappraisal of its pharmacodynamic
and pharmacokinetic properties, and therapeutic efcacy, Drugs (1988), https://
doi.org/10.2165/00003495-198835030-00004.
[28] R.N. Brogden, R.C. Heel, G.E. Pakes, T.M. Speight, G.S. Avery, Diclofenac sodium: a
review of its pharmacological properties and therapeutic use in rheumatic diseases
and pain of varying origin, Drugs (1980), https://doi.org/10.2165/00003495-
198020010-00002.
[29] R. Altman, B. Bosch, K. Brune, P. Patrignani, C. Young, Advances in NSAID
development: evolution of diclofenac products using pharmaceutical technology,
Drugs (2015), https://doi.org/10.1007/s40265-015-0392-z.
[30] M. Tubic-Grozdanis, M.B. Bolger, P. Langguth, Application of gastrointestinal
simulation for extensions for biowaivers of highly permeable compounds, AAPS J.
(2008), https://doi.org/10.1208/s12248-008-9023-x.
[31] B. Jeganathan, V. Prakya, A. Deshmukh, Preparation and evaluation of diclofenac
sodium tablet coated with polyelectrolyte multilayer lm using hypromellose
acetate succinate and polymethacrylates for pH-dependent, modied release drug
delivery, AAPS PharmSciTech (2016), https://doi.org/10.1208/s12249-015-0385-
y.
[32] J. Van Den Abeele, J. Brouwers, R. Mattheus, J. Tack, P. Augustijns,
Gastrointestinal behavior of weakly acidic BCS Class II drugs in man - case study of
diclofenac potassium, J. Pharm. Sci. (2016), https://doi.org/10.1002/jps.24647.
[33] Q. Wang, N. Fotaki, Y. Mao, Biorelevant dissolution: methodology and application
in drug development, Dissolution Technol. (2009), https://doi.org/10.14227/
DT160309P6.
[34] G. Garbacz, B. Kołodziej, M. Koziolek, W. Weitschies, S. Klein, A dynamic system
for the simulation of fasting luminal pH-gradients using hydrogen carbonate
buffers for dissolution testing of ionisable compounds, Eur. J. Pharmaceut. Sci. 51
(2014), https://doi.org/10.1016/j.ejps.2013.09.020.
[35] H.M. Fadda, H.A. Merchant, B.T. Arafat, A.W. Basit, Physiological bicarbonate
buffers: stabilisation and use as dissolution media for modied release systems, Int.
J. Pharm. (2009), https://doi.org/10.1016/j.ijpharm.2009.08.003.
[36] F. Liu, H.A. Merchant, R.P. Kulkarni, M. Alkademi, A.W. Basit, Evolution of a
physiological pH 6.8 bicarbonate buffer system: application to the dissolution
testing of enteric coated products, Eur. J. Pharm. Biopharm. 78 (2011) 151157,
https://doi.org/10.1016/j.ejpb.2011.01.001.
[37] M. Babayeva, A.A. Marfo, R. Wolfe, Y.C.S. Hlaing, Z.G. Loewy, B.A. Selvi, Studying
the factors that impact the dissolution characteristic of complex drug product,
Pharmaceut. Dev. Technol. 24 (2019), https://doi.org/10.1080/
10837450.2019.1647544.
[38] I. Partheniadis, P. Gkogkou, N. Kantiranis, I. Nikolakakis, Modulation of the release
of a non-interacting low solubility drug from chitosan pellets using different pellet
size, composition and numerical optimization, Pharmaceutics 11 (2019), https://
doi.org/10.3390/pharmaceutics11040175.
[39] S. Abdul, A.V. Chandewar, S.B. Jaiswal, A exible technology for modied-release
drugs: multiple-unit pellet system (MUPS), J. Contr. Release 147 (2010), https://
doi.org/10.1016/j.jconrel.2010.05.014.
[40] T.N. Hiew, Y.H. Tian, H.M. Tan, P.W.S. Heng, A mechanistic understanding of
compression damage to the dissolubility of coated pellets in tablets, Eur. J. Pharm.
Biopharm. 146 (2020), https://doi.org/10.1016/j.ejpb.2019.11.006.
[41] S.R. B´
echard, J.C. Leroux, Coated pelletized dosage form: effect of compaction on
drug release, Drug Dev. Ind. Pharm. 18 (1992), https://doi.org/10.3109/
03639049209052410.
D. Zakowiecki et al.
... Limited friability of starter pellets is vital because it prevents the undesired dust formation, especially at the beginning of the coating process. A summary of DCPA pellets characteristics in comparison with other starter pellets can be found elsewhere [9,22,23]. ...
... In this case, it may have been the result of a larger contact area between the layer comprising RSC and the surface of the smaller sized pellets (inert cores). It should be kept in mind that these pellets consist of 80% DCPA [9,22,23], and DCPA consists of about 29.5% w/w calcium, which can stabilize statins. Summarizing the results of studies with various vehicles, the data suggest that kefir appears to be the most favorable for administration of sprinkle IR MUPS formulation with RSC as the drug substance. ...
Article
Full-text available
Sprinkle formulations represent an interesting concept of medicinal products aimed at the steadily growing population of patients suffering from swallowing difficulties (dysphagia). In the present work, immediate-release sprinkle MUPS (multiple-unit pellet system) containing rosuvastatin calcium as a model drug substance was successfully developed. The formulation was prepared by drug layering technique using novel calcium phosphate-based starting pellets (PharSQ® Spheres CM) of three different particle sizes. The study showed that the developed multiparticulates were characterized by uniform distribution of coating layers thickness, as well as fast dissolution rate (more than 85% of rosuvastatin calcium dissolved within 30 min, as required by the relevant USP/NF monograph). Rosuvastatin calcium, like other statins, has a bitter, unpleasant taste. Investigations conducted with an electronic tongue suggested that the developed formulation achieved the desired taste-masking efficiency. The effect was found to be particle size-dependent, improving as the size of the multiparticulates increased.
... Common inert excipients include sugar (sucrose maximum of 92%), sucrose, microcrystalline cellulose (MCC, Cellets, Celphere), which are water insoluble and have high abrasion resistance, making them easy to coat [11,12], starch (Suglets, Nu-Pareil PG) [8,13,14], isomalt, anhydrous dibasic calcium phosphate, lactose, tartaric acid, or siliconbased materials and waxes can be used for layering [15,16]. It is found that the yield and some characteristics of resulting pellets are influenced by the original non-pareil properties such as solubility, hardness, and friability. ...
Article
Full-text available
Pelletization turns fine powders or granules into small, round pellets for easy flow and handling. These pellets offer advantages such as improved absorption, modified drug release patterns & reduced irritation at application sites. Dry powder layering is gaining popularity in the pharmaceutical industry due to its cost and time efficiency. This method is an effective and straightforward process, can be alternative to liquid-based coating techniques. In recent years, active pharmaceutical ingredient layering onto multi-particulate core materials. One innovative pelletization method uses a rotor processor for powder layering. This process involves applying drug powder onto sugar spheres & spraying a binder solution to achieve uniform distribution and enhance drying efficiency. Compared to traditional methods, this dry powder layering is faster & more energy-efficient, reducing drying time & energy consumption. It is a streamlined and eco-friendly method as no solvent is required which makes it beneficial for pellet coating. This article provide insights on the powder layering technique with innovative fluid bed pelletizing technologies like rotor processor, glatt powder coater granulator series, tangential spray in the rotary insert, complex perfect sphere insert and glatt procell series & their applications.
... This study employed a relatively new product, dibasic calcium phosphate (DCPA) starter pellets. While not yet widely recognized in pharmaceutical technology, DCPA pellets offer several promising advantages [32][33][34]. ...
Article
Full-text available
Background/Objectives: The aim of the present study was to develop lactose-free formulations of rivaroxaban, a novel oral anticoagulant used for the treatment and prevention of blood clotting. As a BCS Class II drug, rivaroxaban is characterized by poor solubility in aqueous media, posing a significant formulation challenge. Methods: To address this, phosphate-based excipients were employed to prepare both traditional single-unit dosage forms (tablets) and modern multiple-unit pellet systems (MUPS). These formulations were successfully developed and thoroughly evaluated for their physical properties and performance. Results: The resulting formulations demonstrated very good mechanical strength, including appropriate hardness and friability, alongside strong chemical stability. Their dissolution profiles met the requirements of the compendial monograph for Rivaroxaban Tablets and were comparable to those of the reference product, Xarelto® film-coated tablets. Conclusions: This study shows the potential for producing effective, stable, and patient-friendly medications that meet the needs of contemporary society, where an increasing number of individuals suffer from lactose intolerance or seek vegan-friendly alternatives.
... The coating thickness and osmogens ability to create osmotic pressure impacts on lag time and dissolving rates. [52] Monolithic osmotic system ...
Article
Full-text available
Oral route is one of the most popular and successful delivery routes for the medicines. It achieved a prominent place in drug delivery due to ease of administration by all age groups. The major disadvantage of oral conventional drug therapy is that, it has to be taken 2-3 times a day, and if patient misses any dose it leads to incomplete prescribed dose and delays the time of cure. The considerable efforts have been taken to improve the success rate of oral drug delivery by modifying various parameters related to drug release. Osmosis is the movement of a substance against concentration gradient; in this instance, osmotic delivery use an osmotic gradient to successfully distribute the drug in controlled way and with zero order kinetics and emerging as the most dependable method of regulated drug delivery. The medication release from device is mostly unaffected by stomach secretions. Drug, release control polymer, osmotic agent, semipermeable membrane, and delivery orifice are the major parts of osmotic delivery system. This review mainly focuses on an osmotic drug delivery system providing detailed information regarding its design, different types, and responsible ingredients for controlled release by the virtue of osmosis.
... Te sampled solutions were analyzed at 276 nm using UV-vis spectrophotometer (Merck, Darmstadt, Germany) after they have been fltered. Te amounts of diclofenac sodium present were subsequently calculated[24]. Triplicate determinations were carried out for each capsule formulation.2.9.5. Drug Release Kinetics.Te obtained data from the in vitro diclofenac sodium release studies were integrated into the mathematical kinetic models (zero order, Higuchi, Korsmeyer-Peppas, frst order, and Hixson-Crowell) to determine the mechanisms of diclofenac sodium release from the pectin-clay matrices. ...
Article
Full-text available
A combination of inorganic and organic hybrid systems is of high research interest as they provide novel hybrid systems for the improvement of existing properties, overcoming limitations of the parent materials, and for the optimization of their controlled release potential. This study sorted to develop and pharmaceutically assess the release profile of diclofenac sodium using cocoa pod husk (CPH) blended with different proportions of either talc or bentonite as multiparticulate composite release modifiers. Preformulation investigations of the multiparticulate hybrid systems included pH, swelling index, moisture content, elemental contents, and flow properties. The FTIR was also used to investigate the compatibilities between pectin and bentonite (PB), pectin and talc (PT), and diclofenac and pectin-talc (DPT), as well as diclofenac and pectin-bentonite (DPB). The diclofenac content, uniformity of the weight of capsules, in vitro drug release, and the kinetics and mechanism of release of diclofenac from the hybrid systems were also investigated using mathematical models. The pectin yield was 23.3%, with the water-holding capacities of pectin-talc (PT) and pectin-bentonite (PB) hybrid systems being 6.4% and 5.0%, respectively. The swelling indices of PT and PB were 110.0 and 130.0 in 0.1 M HCL at pH 1.2 and 130.0 and 149.0 in phosphate buffer at pH 6.8, respectively. This system was also found to exhibit excellent flow properties, and there were no diclofenac-excipient interactions. All formulated batches passed the pharmacopoeial and nonpharmacopoeial tests. They also demonstrated controlled release properties via different release kinetics and mechanisms. This study shows that the pectin-talc and pectin-bentonite multiparticulate composites could be used as release modifiers in pharmaceutical preparations.
... It has a short plasma half-life of approximately one to two hours [11]. Various formulations of diclofenac are used for the drug products available in the market or described in the literature [12][13][14][15], including enteric-coated tablets and encapsulated enteric-coated pellets. In this particular case, the capsules are only applied for easier administration and do not have any protective function. ...
Article
Full-text available
Developing delayed-release formulations for acid-sensitive actives can be a costly and time-consuming process. However, ready-to-fill functional capsules, such as EUDRACAP® can significantly mitigate these challenges. The in vitro performance of EUDRACAP® enteric was evaluated in two typical delayed-release scenarios: for diclofenac (a drug that can cause irritation to gastric mucosa), and for omeprazole (a drug susceptible to degradation due to the acidity of gastric fluid). The prototypes were tested in HCl 0.1N according to the USP <711> for at least 2 h and compared to commercial products. The results showed that the performance of EUDRACAP® was below LOD and in compliance with the requirements for drug release in acidic media (NMT 10%). Additionally, the impurities were evaluated after the acidic stress. The low total percentage of impurities of 0.44% for diclofenac (NMT 1.50%) and 0.22% for omeprazole (NMT 2.00%) indicates a very good protection by EUDRACAP®. A comprehensive comparative analysis of the in vitro performance clearly showed the acid protection capability of EUDRACAP® enteric capsules making them a serious alternative to existing enteric dosage forms alternatives. EUDRACAP® is an accessible solution both in large-scale industrial and smaller pharmacy settings. Offering increased accessibility, affordability, and convenience to manufacturers and consumers alike and leading to improved healthcare outcomes.
... Vì mục đích này, các polyme bao tan trong ruột nhạy cảm với pH thường được sử dụng. Các polyme này phải có tác dụng hạn chế sự giải phóng thuốc trong điều kiện acid của dạ dày và cho phép thuốc hòa tan nhanh chóng trong tá tràng và các phần xa của đường tiêu hóa [2]. ...
Article
Background: Diclofenac sodium (DS) is a non-steroidal anti-inflammatory drug (NSAIDs) that has strong anti-inflammatory and analgesic effects, but it has many serious side effects on the gastrointestinal tract. Enteric-coated DS pellets have many advantages such as limited drug release in the stomach, with Eudragit L100 coating to help drug release in the intestines, avoiding side effects in the gastrointestinal tract. The aim of this study was to formulate enteric-coated pellets and to evaluate the physico-chemical properties of prepared pellets. Materials and methods: DS, HPMC, Eudragit L100 were used in this study. DS pellets were prepared using sugar spheres layered with DS followed by an enteric coat of Eudragit L100. Results: Prepared enteric-coated DS pellets by layering method using HPMC as a binding agent and Eudragit L100 as enteric-coated membrane to help pellets ensure the release of drug in the gastrointestinal tract. Pellets have a nice shape, the size range of 0.7-1 mm, the layering efficiency of 36.69%, the drug content of 8.84% ± 0.43% (w/w). The drug release was 3.59% after 2h in acidic environment of pH 1.2 and 84.9% after 1h in phosphate buffered saline of pH 6.8. Conclusions: Enteric-coated pellets of DS were successfully prepared by using coating technology. Key words: diclofenac sodium, pellet, coating technology
Article
Microparticles have unique benefits in the formulation of multiparticulate and multi-unit type pharmaceutical dosage forms allowing improved drug safety and efficacy with favorable pharmacokinetics and patient centricity. On the other hand, the above advantages are served by high and well reproducible quality attributes of the medicinal product where even flexible design and controlled processability offer success as well as possible longer product life-cycle for the manufacturers. Moreover, the specific demands of patients can be taken into account, including simplified dosing regimens, flexible dosage, drug combinations, palatability, and ease of swallowing. In the more than 70 years since the first modified-release formulation appeared on the market, many new formulations have been marketed and many publications have appeared in the literature. More unique and newer pharmaceutical technologies and excipients have become available for producing tailor-made particles with micrometer dimensions and beyond. All these have contributed to the fact that the sub-units (e.g. minitablets, pellets, microspheres) that make up a multiparticulate system can vary widely in composition and properties. Some units have mucoadhesive properties and others can float to contribute to a suitable release profile that can be designed for the multiparticulate formula as a whole. Nowadays, there are some available formulations on the market, which are able to release the active substance even for several months (3 or 6 months depending on the type of treatment). In this review, the latest developments in technologies that have been used for a long time are presented, as well as innovative solutions such as the applicability of 3D printing to produce subunits of multiparticulate systems. Furthermore, the diversity of multiparticulate systems, different routes of administration are also presented, touching the ones which are capable of carrying the active substance as well as the relevant, commercially available multiparticle-based medical devices. The versatility in size from 1 µm and multiplicity of formulation technologies promise a solid foundation for the future applications of dosage form design and development.
Article
Full-text available
This article presents various types of starter pellets (inert cores containing no drug substance), which are frequently used for the production of MDDS. Their physicochemical and functional properties, which normally are not part of the specification, may prove to be important factors determining the course of the drug layering process as well as the properties of the finished drug product. It is worth giving them some consideration before starting the development works.
Article
Full-text available
Two size classes of piroxicam (PXC) pellets (mini (380–550 μm) and conventional (700–1200 μm)) were prepared using extrusion/spheronization and medium viscosity chitosan (CHS). Mixture experimental design and numerical optimization were applied to distinguish formulations producing high sphericity pellets with fast or extended release. High CHS content required greater wetting liquid volume for pellet formation and the diameter decreased linearly with volume. Sphericity increased with CHS for low-to-medium drug content. Application of PXRD showed that the drug was a mixture of form II and I. Crystallinity decreased due to processing and was significant at 5% drug content. Raman spectroscopy showed no interactions. At pH 1.2, the dissolved CHS increased ‘apparent’ drug solubility up to 0.24 mg/mL while, at pH 5.6, the suspended CHS increased ‘apparent’ solubility to 0.16 mg/mL. Release at pH 1.2 was fast for formulations with intermediate CHS and drug levels. At pH 5.6, conventional pellets showed incomplete release while mini pellets with a CHS/drug ratio ≥2 and up to 21.25% drug, showed an extended release that was completed within 8 h. Numerical optimization provided optimal formulations for fast release at pH 1.2 with drug levels up to 40% as well as for extended release formulations with drug levels of 5% and 10%. The Weibull model described the release kinetics indicating complex or combined release (parameter ‘b’ > 0.75) for release at pH 1.2, and normal diffusion for the mini pellets at pH 5.6 (‘b’ from 0.63 to 0.73). The above results were attributed mainly to the different pellet sizes and the extensive dissolution/erosion of the gel matrix was observed at pH 1.2 but not at pH 5.6.
Article
Full-text available
Oral modified-release multiparticulate dosage forms, which are also referred to as oral multiple-unit particulate systems, are becoming increasingly popular for oral drug delivery applications. The compaction of polymer-coated multiparticulates into tablets to produce a sustained-release dosage form is preferred over hard gelatin capsules. Moreover, multiparticulate tablets are a promising solution to chronic conditions, patients' adherence, and swallowing difficulties if incorporated into orodispersible matrices. Nonetheless, the compaction of multiparticulates often damages the functional polymer coat, which results in a rapid release of the drug substance and the subsequent loss of sustained-release properties. This review brings to the forefront key formulation variables that are likely to influence the compaction of coated multiparticulates into sustained-release tablets. It focusses on the tabletting of coated drug-loaded pellets, microparticles, and nanoparticles with a designated section on each. Furthermore, it explores the various approaches that are used to evaluate the compaction behaviour of particulate systems.
Article
Damage to the drug diffusion coat barrier of controlled release pellets by the compaction force when preparing multiple-unit pellet system tablets is a major concern. Previous studies have shown that pellets located at the tablet axial and radial peripheral surfaces were more susceptible to damage when compacted due to the considerable shear encountered at these locations. Hence, this study was designed to assess with precision the impact of pellet spatial position in the compact on the extent of coat damage by the compaction force via a single pellet in minitablet (SPIM) system. Microcrystalline cellulose (MCC) pellet cores were consecutively coated with a drug layer followed by a sustained-release layer. Chlorpheniramine maleate was the model drug used. Using a compaction simulator, the coated pellets were compacted singly into 3 mm diameter SPIMs with MCC as the filler. SPIMs with individual pellets placed in seven positions were prepared. The uncompacted and compacted coated pellets, as SPIMs, were subjected to drug release testing. The dissolution results showed that pellets placed at the top-radial position were the most susceptible to coat damage by the compaction force, while pellets positioned within the minitablet at the middle and upper quadrant positions showed the least damage. The SPIM system was found to be effective at defining the extent of coat damage to the pellet spatial position in the compact. This study confirmed that coated pellets located at the periphery were more susceptible to damage by compaction, with pellets located at the top-radial position showing the greatest extent of coat damage. However, if the pellet was completely encrusted by the cushioning filler, coat damage could be mitigated. Further investigations were directed at how the extent of coat damage impacted drug release. Interestingly, small punctures were found to be most detrimental to drug release whilst coats with large surface cuts did not completely fail. A damaged pellet coat has some self-sealing ability and failure is not total. Thus, this study provides a deeper understanding of the consequence of coat damage to drug release when sustained release coated pellets are breached.
Article
This article summarizes the critical factors involved in product development of a single dosage form formulated by compacting ethyl cellulose (EC) coated controlled release pellets into a tablet. The greatest challenge associated with this type of complex system is to minimize the effect of compression on the drug release. The effects of compression on the drug release were optimized with combination of the following factors (1) particle size of the core pellets, (2) the selection of the coating polymer’s viscosity grade, and (3) emergence of cushioning agents. The optimization of these factors provided superior protection for the controlled release coated pellets; therefore, the desired drug release from the tablet was successfully achieved as designed. However, the drug release rates from the coated pellets before and after the compression were minimized and exhibited only a slight difference.
Chapter
The aim of this chapter is to provide a comprehensive overview of oral controlled release (CR) technology, based, to a large extent, on the author's experience and expertise from leading a CR centre of excellence for a multinational large pharma company and the internal and external collaborations or interactions that came with that role. The chapter discusses key considerations for CR formulation development, namely (i) the gastrointestinal tract, active pharmaceutical ingredient (API) attributes and manufacturability/quality by design (QbD), (ii) key process and formulation technologies and (iii) in vitro and in vivo evaluation of CR dosage forms. CR formulation technologies discussed are (i) hydrophilic matrix tablets, (ii) inert matrices, (iii) multiparticulate technologies, namely extrusion-spheronisation, direct pelletisation, dry powder layering, spray layering, hot-melt pelletisation, mini-tablets and multiple unit pellet system (MUPS) tablets, (iv) osmotic drug delivery systems and (v) proprietary and other technologies, including so-called diagnostic tools (i.e. Enterion™, IntelliCap®) and gastroretention technologies. While a single chapter can never provide universally applicable recipes or decision trees, it will hopefully give formulators a good overview to guide their own work in this exciting area of pharmaceutical formulation development.
Article
The tablet of multi-unit pellet system (TMUPS), using coated pellets, for controlled release of drugs is an effective therapeutic alternative to conventional immediate-release dosage forms. The main advantages of TMUPS include a) ease of swallowing and b) divisible without compromising the drug release characteristics of the individual units. TMUPS can be prepared more economically than pellet-filled capsules because of the much higher production rate of tableting process. In spite of the superiorities of TMUPS, its adoption has been challenged by manufacturing problems, such as compromised integrity of coated pellets and poor content uniformity. Herein, we provide an updated review on research, from both scientific literatures and patents, related to the compaction of TMUPS. Factors important for the successful production of TMUPS are summarized, including model drug property, potential cushioning agents, and novel techniques to protect pellets from damage. This review is intended to facilitate the future development of manufacturable TMUPS with drug release behavior similar to that of the original coated pellets.
Chapter
For many multiparticulate products, the process begins with an inert core. As the starting material, the characteristics of an inert core influence each successive step including the end-product performance. Identifying the critical to quality attributes (CQA) of an inert core and how they influence a product is essential throughout the development, scale-up, and manufacturing stages. In this chapter, various characteristics such as surface area, particle size distribution, various density, shape, surface morphology, robustness and processability, hardness and tensile strength, and friability are discussed. These tests are beyond the pharmacopeial tests of standard and purity and usually do not appear on most of the inert core excipient manufacturers’ certificate of analysis. Understanding these characteristics helps in developing a robust product and also understands any unforeseen variability between different and the same batch of final multiparticulate dosage form.
Article
The present time is considered as an era of advancements in drug delivery systems. Different nove approaches are under investigation that range from uniparticulate to multi particulate system, macro to micr and nano particulate systems. Pelletization is one of the novel drug delivery technique that provides an effectiv way to deliver the drug in modified pattern. It is advantageous in providing site specific delivery of th drug. Drugs with unpleasant taste, poor bioavailability and short biological half-life can be delivered efficientl through pellets. Their reduced size makes them more valuable as compared to the conventional drug deliver system. Different techniques are used to fabricate the pellets such as extrusion and spheronization, hot mel extrusion, powder layering, suspension or solution layering, freeze pelletization and pelletization by direct compressio method. Various natural polymers including xanthan gum, guar gum, tragacanth and gum acacia, semisyntheti polymers like cellulose derivatives, synthetic polymers like derivatives of acrylamides, can be used i pellets formulation. Information provided in this review is collected from various national and internationa research articles, review articles and literature available in the books. The purpose of the current review is t discuss pellets, their characterizations, different techniques of pelletization and the polymers with potential o being suitable for pellets formulation.