You are on page 1of 8

J Therm Anal Calorim (2016) 123:2283–2290

DOI 10.1007/s10973-015-4935-z

The effect of Pluronic F127 on the physicochemical properties


and dissolution profile of lovastatin solid dispersions
Bo_zena Karolewicz1 • Maciej Gajda1 • Janusz Pluta1 • Agata Górniak2

Received: 28 April 2015 / Accepted: 16 July 2015 / Published online: 26 July 2015
 Akadémiai Kiadó, Budapest, Hungary 2015

Abstract Lovastatin (LOV) is a BCS class II drug and respectively, into a hard gelatin capsule was higher (100.0,
hence has poor water solubility, due to which its rate of 83.37, 98.95 and 87.50 %, respectively) than that of pure
solubilization is low. The objective of this study was to LOV (71.15 %) within 30 min.
improve the dissolution rate of LOV by preparing, using the
kneading method, solid dispersions with Pluronic F127 Keywords Lovastatin  Pluronic F127  Solid dispersion 
(PLU) as a carrier. LOV/PLU solid dispersions were char- DSC  Phase diagram  Intrinsic dissolution rate
acterized by means of differential scanning calorimetry
(DSC), Fourier transform infrared spectroscopy (FTIR),
X-ray powder diffractometry (XRPD), and in vitro dissolu- Introduction
tion tests. The DSC investigation revealed that LOV and
PLU form a simple eutectic system containing 6.0 % w/w of Several reports in the literature indicate the importance of the
LOV at the eutectic point. FTIR spectroscopy and XRPD substance dissolution process for its absorption from the
studies indicated no interaction between LOV and PLU in gastrointestinal tract and its later therapeutic efficiency. The
the solid state. The intrinsic dissolution rate and in vitro low aqueous solubility of LOV (4 9 10-4 mg mL-1), in
release into a hard gelatin capsule of LOV from solid dis- addition to its first pass metabolism, results in a low oral
persions was compared to pure LOV. The dissolution rate of bioavailability (less than 5 %) [1]. Achieving the high disso-
LOV reached within 24 h was improved among all solid lution rate of lipophilic drugs such as LOV is a key aim in the
dispersions with PLU. The intrinsic dissolution rate in pH 7.0 development of formulations with rapid dissolution and
phosphate buffer with 0.5 % sodium lauryl sulfate of LOV in improved bioavailability, as they will simultaneously improve
solid dispersion containing the substance by mass percentage patient compliance and ultimately reduce the total therapeutic
of 60, 70 and 80 % w/w increased more than 30-fold. While dose of the drug. Various techniques have been applied when
within 24 h 16.53 % of the pure drug was dissolved, 100 % attempting to improve LOV dissolution rate [2], including
of the drug was dissolved with the formulations containing obtaining solid lipid nanoparticles [3], liquisolid systems [4],
50–80 % w/w of LOV within 240 min. The dissolution rate self-microemulsifying drug delivery systems [1, 5], the for-
of LOV from solid dispersions 50/50 LOV/PLU, 60/40 LOV/ mation of water soluble complexes and other methods by the
PLU, 70/30 LOV/PLU and 80/20 LOV/PLU percent, use of superdisintegrants [6], solid dispersions [7, 8], surfac-
tants [9] and particle size reduction [10]. The release of LOV
from solid dosage forms can be enhanced by the addition of
& Bo_zena Karolewicz suitable super disintegrants; however, this does not improve
bozena.karolewicz@umed.wroc.pl its solubility and does not decrease the size of the particles, so
1 the substance is still poorly wetted [6]. Solid dispersions
Department of Drug Form Technology, Wroclaw Medical
University, Borowska 211A, 50-556 Wroclaw, Poland combine the benefits of a local increase in the solubility
2 (within the solid solution) with maximizing the surface area of
Laboratory of Elemental Analysis and Structural Research,
Wroclaw Medical University, Borowska 211A, the compound that comes into contact with the dissolution
50-556 Wroclaw, Poland medium as the carrier dissolves [6].

123
2284 B. Karolewicz et al.

There are a number of carriers used in the preparation of in hard gelatin capsule were performed in pH 7.0 phosphate
LOV solid dispersions like as polymers such as Poloxamer buffer with 0.5 % SLS and compared with pure LOV.
F-68 [11], polyethylene glycol 4000 and 6000 (PEG 4000,
PEG 6000), polyvinylpyrrolidone K30 (PVP K30) [7], sodium
starch glycolate, croscarmellose sodium, crospovidone Experimental
[6, 12], surfactants, or others, e.g., water/tertiary butyl alcohol
(TBA) with mannitol [13]. Materials
The in vitro LOV release from tablets containing solid
dispersion prepared by solvent evaporation method using Samples of LOV (99 % purity) was obtained as a gift from
Poloxamer F-68 showed a faster release of the substance in Polpharma S.A. (Poland). PLU was supplied from Sigma-
comparison to tablets with pure drug and marketed formu- Aldrich (USA). Sodium lauryl sulfate (SLS) was purchased
lations [11]. Patel et al. used a modified locust bean gum to from P.P.H. ‘‘Stanlab’’ (Poland). Ethanol and acetonitrile
prepare LOV solid dispersions and achieved an enhance- HPLC grade were obtained from Fluka Biochemica (Ger-
ment in the dissolution rate of substance. An in vivo study of many). Phosphoric acid was purchased from Fluka Bio-
the inhibition activity of 3-hydroxy-3-methyl-glutaryl- chemica (Germany). Sodium hydrogen phosphate,
coenzyme A reductase showed a significant reduction in its potassium dihydrogen phosphate, and phosphoric acid
activity in the case of solid dispersions of LOV [8]. Solid were purchased from Chempur (Poland).
dispersions were also prepared by using PEG 4000 and PVP
K30 through a fusion-cooling and solvent evaporation Preparation of solid dispersion
technique. Tablets containing these solid dispersions showed
a significant improvement in the release profile of LOV Accurately weighed amounts of LOV were mixed with
compared with tablets containing LOV without polymers different concentrations of in agate mortar, while a suffi-
[7]. Raja Rajeswari et al., using the hot melt extrusion cient volume of ethanol was added to achieve a consistency
technique, prepared LOV solid dispersions with Soluplus like slurry. The solvent was then completely evaporated at
(polyvinyl caprolactam-polyvinyl acetate-polyethylene gly- 40–45 C while being continuously stirred to obtain a dry
col graft copolymer) where the drug was in an amorphous mass. The obtained solid dispersions were pulverized
state in the molten polymer. All the formulations showed a powders, sieved using a 315-lm sieve and then stored in a
marked increase in the drug release when there was an desiccator at room temperature until use. The mass ratios
increase in the concentration of Soluplus [9]. In studies by of the LOV/PLU mixtures were 90/10, 80/20, 70/30, 60/30,
Verma et al. [13], an increase in the solubility and dissolu- 50/50, 40/60, 30/70, 20/80 and 10/90 percent, respectively.
tion rate was obtained for solid dispersion (7:5 ratio of TBA/
water) prepared by freeze–dried method due to the conver- Drug content
sion of the crystalline nature of the drug an amorphous one,
as well as a decrease in the particle size of the drug. The equivalent mass of solid dispersions containing 10 mg
Pluronic F127, also known as Poloxamer 407, belongs to of LOV was weighed accurately and dissolved in 10 mL of
the category of nonionic surface-active triblock copolymer acetonitrile. The solution was filtered, and LOV content
consisting of a central hydrophobic block of polypropylene was analyzed by HPLC.
glycol flanked by two hydrophilic blocks of polyethylene
glycol. This polymer is exploited in drug form technology Differential scanning calorimetry (DSC)
as emulsifier, solubilizer and wetting agent for poor water-
soluble drugs. Drugs from II BSC class may be solubilized The DSC curves of each mixture were obtained using a
with a core of micelle or conjugated to the micelle-forming differential scanning calorimeter (Mettler Toledo DSC 25)
polymers which lead to an increase in dissolution of these equipped with a heat flow sensor and joined via an inter-
substances [14]. face TA Controller TC 15 to a computer. Measurements
The present study examines LOV–PLU solid dispersions were recorded by STARe software. Samples for DSC
prepared in 10/90, 20/80, 30/70, 40/60, 50/50, 60/40, 70/30, measurements were sealed in 40 lL standard aluminum
80/20 and 90/10 mass ratios by kneading method. To crucibles with a single hole punched in the lid. An empty
investigate the properties of the obtained series of solid pan of the same type was employed as a reference. The
dispersions of LOV–PLU, differential scanning calorimetry DSC instrument was calibrated using the melting point of
(DSC), X-ray powder diffraction (XRD) and Fourier trans- indium (156.6 ± 0.3 C) as a standard. DSC scans of each
form infrared spectroscopy (FTIR) methods were employed. mixture were performed at a heating rate of 5 C min-1 at
Intrinsic dissolution studies of LOV from obtained solid a temperature range of 25 to 190 C. The DSC cell was
dispersions and the release substance from solid dispersions purged with a stream of dry argon at a rate of

123
The effect of Pluronic F127 on the physicochemical properties and dissolution profile of… 2285

50 mL min-1. Experiments were performed in triplicate, performed using an HPLC system (System GOLD 126,
and the mean values were calculated. Beckman Coulter) with a UV–VIS detector. The analysis
was carried out with the use of Zorbax SB-C8
Powder X-ray diffraction analysis (XRPD) (250 9 4.6 mm, 5 lm, Agilent). An HPLC analysis was
performed by gradient elution with a flow rate of
Powder X-ray diffraction patterns were recorded on a 1.5 mL min-1. The mobile phase composition was ace-
powder diffractometer (D2 Phaser, Bruker) with CuKa tonitrile–water with 0.1 % phosphoric acid [15]. Substances
radiation with LYNXEYE detector. The degree of eluted from the column were identified by a UV visible
diffractions was measured at 15 min-1 between 5 and detector at 238 nm. External standards of LOV were used to
40 (2h) with an accuracy of 0.02 throughout the mea- obtain calibration curves. The linear correlation coefficient
surement range, at 0.5 s/step. (r2) was greater than 0.99 in the range of 5–285 lg mL-1.

Fourier transform infrared spectroscopy (FTIR) In vitro of LOV dissolving into hard gelatin capsule

FTIR spectra were registered by using a Spectrum Two Release studies were carried out in triplicate in USP
spectrophotometer (PerkinElmer) with a UATR accessory Apparatus 2 (Basket type). Solid dispersions equivalent to
attached. The scanning range was 4000–450 cm-1. The 40 mg of drug in gelatin capsules were added to 500 mL of
spectral resolution for all step-scan FTIR measurements pH 7.0 phosphate buffer with 0.5 % SLS stirred at 75 rpm.
was 4 cm-1. Aliquots of 3 mL were withdrawn at specified time inter-
vals and analyzed by HPLC at 238 nm.
Dissolution studies

Intrinsic dissolution (IDR) studies Results and discussion

The dissolution test was conducted under sink conditions in Drug content
1000 mL of pH 7.0 phosphate buffer with 0.5 % SLS at
37 ± 0.5 C and rotational speeds of 50 rpm. The disso- The LOV content of the formulations was found to be in
lution system was fitted with SR8-PLUS (Hanson) and a the range of 99.21 % to 102.55 % of the declared amount.
7-channel peristaltic pump. LOV (100.0 mg) or an equiv-
alent amount of solid dispersion disks was prepared by Differential scanning calorimetry study
compressing powder in hydraulic press (Specac, Mettler
Toledo) for 1 min under a 1t compression force, using a The DSC curves of pure components, obtained at a heating
8-mm punch. Samples were withdrawn at appropriate time rate of 5 C min-1 in the temperature range from 25 to
intervals. The quantitative determination for LOV was 190 C, are presented in Fig. 1. The DSC curve of pure
Fig. 1 DSC curves of pure
LOV and PLU ^Exo

LOVASTATIN
Integral –512.94 mJ
Normalized –104.68 Jg^–1
Peak 171.75 °C

20 PLURONIC F127
mW
Integral –1606.00 mJ
Normalized –176.29 Jg^–1
Peak 53.42 °C

40 60 80 100 120 140 160 180 °C

0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30 min

123
2286 B. Karolewicz et al.

LOV shows only one endothermic peak corresponding to second peak (the temperature of liquid) was generally
the melting of the drug at 171.8 C, which corresponds wider, indicating that the complete melting took place over
with the data presented by Yoshida et al. [16]. Similarly, a temperature range. Terminal solid solutions were not
the DSC curve of PLU, presented in the same figure, shows observed at either sides of the phase diagram. The melting
one endothermic effect, associated with its melting at points of the pure components (LOV: 171.8 C, PLU:
53.4 C. This confirms that under these conditions both 53.4 C) were depressed due to the existence of the other
components are stable and do not decompose. component in the mixture as shown in the phase diagram in
Figure 2 shows the phase diagram of the examined Fig. 2. The values of the eutectic melting enthalpy DH for a
system constructed on the basis of the DSC results. The given dispersion, determined by integration of the eutectic
DSC curves of obtained dispersions (Fig. 3) clearly indi- peak area on DSC curves, are plotted in Fig. 4 versus mass
cate that the investigated compounds formed a simple fraction of LOV (Tamman’s triangle construction [17]).
binary eutectic system—only two kinds of thermal effects The thermal effect of the eutectic transition goes to zero for
are shown for the whole range of compositions. The onset a composition corresponding to pure LOV. This confirms
of the first peak consistently appeared near 48.0 C that there is no mutual miscibility in the solid state and no
(Fig. 3), indicating a eutectic reaction: solid lovastatin formation of a terminal solid solution. The values of the
(LOV) ? solid Pluronic F127 (PLU) = liquid (L). The

200.0

180.0
200.0
160.0
180.0
L 140.0
160.0
120.0
ΔT/J g–1

140.0
100.0
120.0
T/°C

80.0
L + LOV
100.0 60.0
80.0 40.0

60.0 20.0

40.0 0.0
0.00 10.00 20.00 30.00 40.00 50.00 60.00 70.00 80.00 90.00 100.00
20.0 PLU + L PLU + LOV LOV/mass%

0.0 10.0 20.0 30.0 40.0 50.0 60.0 70.0 80.0 90.0 100.0 Fig. 4 Determination of the eutectic composition in the LOV–PLU
PLU LOV/mass% LOV system by Tamman’s triangle construction for the eutectic melting
enthalpy DH at 48.0 C (filled triangles a) and by plotting noneutectic
Fig. 2 Phase diagram of the LOV–PLU system melting enthalpy versus mass ratio of LOV (filled circles)

Fig. 3 DSC curves of obtained


LOV/PLU solid dispersions 10/90 %w/w LOV/PLU
20/80 %w/w LOV/PLU
30/70 %w/w LOV/PLU
40/60 %w/w LOV/PLU
50/50 %w/w LOV/PLU
60/40 %w/w LOV/PLU
70/30 %w/w LOV/PLU
80/20 %w/w LOV/PLU
90/10 %w/w LOV/PLU
2
–1
Wg^

^Exo 40 60 80 100 120 140 160 180 °C

123
The effect of Pluronic F127 on the physicochemical properties and dissolution profile of… 2287

eutectic melting enthalpy DH (Fig. 4, filled triangles) • Eutectic composition: mass fraction of LOV 6.0 %,
increase linearly with the content of PLU. Near the eutectic mass fraction of PLU 94.0 %;
point, the DSC scans showed a characteristic overlap of • Eutectic temperature: 48.0 C.
two endotherms (eutectic and liquid events) into a single
peak. Because of this, the eutectic composition was
X-ray diffraction
determined by plotting the non-eutectic melting enthalpy
(after the eutectic melting) of LOV as a function of the
The X-ray diffraction patterns of pure LOV, PLU and solid
mass percentage of the drug and extrapolating the fitted
dispersions are presented in Fig. 5. The XRD patterns of
line to zero enthalpy (Fig. 4, filled circles). Finally, the
pure LOV showed a series of peaks, which are character-
parameters of the eutectic point (E) have been established
istic of a crystalline compound at diffraction angles (2h) at
as follows:
6.80, 8.19, 8.86, 9.75, 14.06, 14.38, 14.83, 15.43,
16.28, 16.69, 17.76, 18.87, 20.86, 21.20, 21.87,
23.10 and 26.95. The X-ray diffractograms of pure PLU
show distinct peaks at 18.87 band 23.10. Drug crys-
(k)
tallinity peaks were also detectable in obtained solid dis-
(j)
persions. The diffraction patterns of solid dispersions
(i)
showed the presence of characteristic diffraction peaks of
(h)
Intensity/a.u.

both the drug and polymer, indicating that LOV was pre-
(g)
(f)
sent as a crystalline material and suggesting that these are
(e)
simple mixtures of drug and carrier without any chemical
(d) interaction between the components.
(c)
(b)
(a) Fourier transform infrared spectroscopy
5 10 15 20 25 30 35 40
2θ /° Figure 6 shows FTIR spectra of PLU, LOV and its solid
dispersions. The spectra of the pure drug presented, simi-
Fig. 5 XRPD patterns of: LOV (a), 90/10 % w/w LOV/PLU (b), larly to that found in works [1, 18], characteristic peaks at
80/20 % w/w LOV/PLU (c), 70/30 % w/w LOV/PLU (d), 60/40 % 3539 cm-1 (alcohol OH stretching vibration), at
w/w LOV/PLU (e), 50/50 % w/w LOV/PLU (f), 40/60 % w/w LOV/
PLU (g), 30/70 % w/w LOV/PLU (h), 20/80 % w/w LOV/PLU (i), 3016 cm-1 (olefinic CH stretching vibration), at
10/90 % w/w LOV/PLU (j), PLU (k) 2965 cm-1 (methyl C–H asymmetric stretching), at

Fig. 6 FTIR spectra of: LOV


(a), 90/10 % w/w LOV/PLU (k)
(b), 80/20 % w/w LOV/PLU
(j)
(c), 70/30 % w/w LOV/PLU
(d), 60/40 % w/w LOV/PLU (i)
(e), 50/50 % w/w LOV/PLU (f),
40/60 % w/w LOV/PLU (g), (h)
30/70 % w/w LOV/PLU (h),
(g)
20/80 % w/w LOV/PLU (i),
10/90 % w/w LOV/PLU (j), (f)
PLU (k)
(e)

(d)

(c)

(b)

(a)

4000 3700 3400 3100 2800 2500 2200 1900 1600 1300 1000 700 400
Wavenumber/cm–1

123
2288 B. Karolewicz et al.

2929 cm-1 (methylene C–H asymmetric stretching), at Dissolution studies


2865 cm-1 (methyl and methylene CH asymmetric
stretching vibration), at 1724, 1710 and 1698 cm-1 (lac- The maximum concentration of dissolved LOV reached
tone and ester carbonyl stretch) and at 871 cm-1 (trisub- within 24 h was enhanced for solid dispersions containing
stituted olefinic CH). The FTIR spectrum of PLU is 80/20, 70/30, 60/40, 50/50, 40/60, 30/70 and 20/80 % w/w.
characterized by principal absorption peaks at 2882 cm-1 However, the initial release rate is enhanced for all solid
(C–H stretch aliphatic), 1342 cm-1 (in-plane O–H bend), dispersions, and the saturation level was reached within
1279 cm-1 (CH2 bending) and 1100 cm-1 (C–O stretch), 240 min of the dissolution process for solid dispersions
which were consistent in all binary systems with the drug. 60/40, 70/30 and 80/20 % w/w (see Figs. 7, 8). In Table 1,
This indicates the absence of drug–polymer interactions, as the intrinsic dissolution rate (IDR) of the pure drug and
all the specific peaks of drug were present in the solid solid dispersions LOV/PLU in pH 7.0 phosphate buffer with
dispersion. 0.5 % SLS were collected. Kaplan [19] noted that com-
pounds with an IDR below 0.1 mg cm-2 min-1 usually
exhibited a dissolution rate-limited absorption. The IDR of
pure LOV of 0.023 mg cm-2 min-1 falls into this category.
The intrinsic dissolution rate of LOV in a solid dispersion
110
containing the substance by mass percentage of 60, 70 and
100 80 % w/w increased more than 30-fold. After 240 min of
90 the test, more than 99 % of LOV was released from the
80 10/90LOV/PLU solid dispersion containing 60–80 % w/w of LOV, when at
Drug dissolved/%

20/80LOV/PLU
70 30/70LOV/PLU this time only 1.40 % of pure LOV had been dissolved.
60 40/60LOV/PLU
50/50LOV/PLU
In vitro results of LOV dissolving into a hard gelatin
50 60/40LOV/PLU capsule in pH 7.0 phosphate buffer with 0.5 % SLS are
40 70/30LOV/PLU
80/20LOV/PLU reported in Table 2. The graphical presentation of the
30 90/10LOV/PLU
dissolution profile into hard gelatin capsules of pure LOV,
20 LOV

10
and LOV/PLU solid dispersions over a period of 120 min
0 is shown in Fig. 9. The dissolution rate of LOV from solid
0 2 4 6 8 10 12 14 16 18 20 22 24 dispersions 50/50 LOV/PLU, 60/40 LOV/PLU, 70/30
t /h LOV/PLU and 80/20 LOV/PLU was higher (100.0, 83.37,
Fig. 7 Dissolution profiles of LOV and its solid dispersions with
98.95 and 87.50 %, respectively) than that of pure LOV
PLU within 24 h of dissolution process (71.15 %) within 30 min.

Fig. 8 Dissolved amount of


LOV from solid dispersions 210
Dissolved amount of LOV/mg 0.5 cm–2

180

10/90LOV/PLU
150 20/80LOV/PLU
30/70LOV/PLU
40/60LOV/PLU
120
50/50LOV/PLU
60/40LOV/PLU
90 70/30LOV/PLU
80/20LOV/PLU
90/10LOV/PLU
60
LOV

30

0
0 120 240 360 480 600 720 840 960 1080 1200 1320 1440
t /min

123
The effect of Pluronic F127 on the physicochemical properties and dissolution profile of… 2289

Table 1 Intrinsic dissolution rate (IDR) of pure LOV and prepared solid dispersions and corresponding ratios. Data are expressed as
mean ± SD (n = 3)
Dispersion code IDR/mg cm-2 min-1 r2 IDR ratio SDs/LOV

10/90 LOV/PLU 0.069 ± 0.0012 0.9989 2.97


20/80 LOV/PLU 0.162 ± 0.0014 0.9974 6.98
30/70 LOV/PLU 0.245 ± 0.0061 0.9988 10.56
40/60 LOV/PLU 0.284 ± 0.0002 0.9881 12.28
50/50 LOV/PLU 0.547 ± 0.0061 0.9996 23.63
60/40 LOV/PLU 0.824 ± 0.0265 0.9977 35.58
70/30 LOV/PLU 0.836 ± 0.0049 0.9917 36.12
80/20 LOV/PLU 0.853 ± 0.0022 0.9633 36.82
90/10 LOV/PLU 0.111 ± 0.0161 0.8147 4.80
Pure LOV 0.023 ± 0.0004 0.9995 –

Table 2 Percentage of lovastatin dissolved within 60 min


Dispersion code Q5min Q30min Q60min

10/90 LOV/PLU 0.71 ± 0.26 1.30 ± 1.99 11.75 ± 5.39


20/80 LOV/PLU 0.04 ± 0.02 2.15 ± 0.53 21.75 ± 3.82
30/70 LOV/PLU 2.47 ± 1.70 79.64 ± 6.30 99.36 ± 3.19
40/60 LOV/PLU 0.77 ± 0.71 10.13 ± 9.15 90.63 ± 8.26
50/50 LOV/PLU 25.42 ± 9.96 100.32 ± 7.12 100.57 ± 0.46
60/40 LOV/PLU 0.25 ± 0.07 83.37 ± 15.94 100.75 ± 5.49
70/30 LOV/PLU 16.67 ± 9.32 98.95 ± 7.46 100.08 ± 5.515
80/20 LOV/PLU 1.127 ± 0.17 87.50 ± 0.60 97.20 ± 1.25
90/10 LOV/PLU 0.63 ± 0.53 70.95 ± 5.34 96.66 ± 0.38
Pure LOV 2.22 4 ± 0.39 71.15 ± 7.30 96.51 ± 2.11
Q5min, Q30min and Q60min values (percent drug dissolved within 5, 30 and 60 min, respectively). Mean ± SD, n = 3

110 Conclusions
100
90 The presented DSC studies demonstrate that LOV and PLU
Cululative drug release/%

10/90LOV/PLU
80 20/80LOV/PLU formed a simple eutectic phase diagram. The FTIR spec-
70 30/70LOV/PLU
40/60LOV/PLU
troscopy and XRPD studies of the obtained dispersions
60
50/50LOV/PLU showed no interaction between the components in the solid
50
60/40LOV/PLU state and confirmed the absence of terminal solid solutions.
40 70/30LOV/PLU
30
Our study demonstrates the successful application of
80/20LOV/PLU

20 90/10LOV/PLU
PLU for the dissolution enhancement of the poorly water-
10 LOV soluble drug LOV. The dissolution rate of LOV was
0 enhanced for all investigated dispersions. It was shown that
0 20 40 60 80 100 120
the release profile of the solid dispersions is strongly
t /min dependent on the content of the polymeric carrier. How-
Fig. 9 Lovastatin dissolving from solid dispersions contained into ever, the initial release rate from solid dispersions con-
hard gelatin capsules within 120 min taining 50–80 % w/w LOV was drastically increased due to

123
2290 B. Karolewicz et al.

a large dissolution rate in these formulations. The results of 9. Raja Rajeswari K, Abbulu K, Sudhakar M, Vishwanadhami M,
this work suggest that the use of PLU as a polymer carrier Tejaswini MS. Studies on dissolution enhancement of lovastatin
using soluplus by solid dispersion technique. Int J Pharm Pharm
for LOV in solid dispersions, as in the case of simvastatin Sci. 2012;4:124–8.
[20], could be a promising approach to improve its release 10. Nanjwade Basavaraj K, Derkar GK, Bechra HM, Nanjwade VK,
from oral form and hence may affect its absorption rate. Manvi FV. Design and characterization of nanocrystals of
lovastatin for solubility and dissolution enhancement.
Acknowledgements The authors would like to thank Polpharma S. J Nanomedic Nanotechnol. 2011;2:2–7.
A. for providing lovastatin. 11. Katare MK, Kohli S, Jain AP. Evaluation of dissolution
enhancement of lovastatin by solid dispersion technique. Int J
Pharm Life Sci. 2011;2:894–8.
12. Shaikh K, Patwekar S, Payghan S, D’Souza J. Dissolution and
References stability enhancement of poorly water soluble drug—Lovastatin
by preparing solid dispersions. Asian J Biomed Pharm Sci.
1. Goyal U, Arora R, Aggarwal G. Formulation design and evalu- 2011;1:24–31.
ation of a self-microemulsifying drug delivery system of lovas- 13. Verma U, Naik JB, Mokale VJ. Preparation of freeze-dried solid
tatin. Acta Pharm. 2012;62:357–70. dispersion powder using mannitol to enhance solubility of
2. Neduri K, Bonthai VK, Vemula SK. Different techniques to Lovastatin and development of sustained release tablet dosage
enhance the dissolution rate of lovastatin: formulation and eval- form. AJPSN. 2014;1:11–26.
uation. Asian J Pharm Clin Res. 2013;6:56–60. 14. Dumortier G, Grossiord JL, Agnely F, Chaumeil JC. A review of
3. Suresh G, Manjunath K, Venkateswarlu B, Satyanarayana V. poloxamer 407 pharmaceutical and pharmacological character-
Preparation, characterization, and in vitro and in vivo evaluation istics. Pharm Res. 2006;23:2709–28.
of Lovastatin solid lipid nanoparticles. AAPS Pharm Sci Tech. 15. European Pharmacopoeia (Ph. Eur.) 8th edn, European Union,
2007;8:162–70. 2014.
4. Shyam J, Ravi KV, Madhavi K, Vamshi KM, Sudheer KD. 16. Yoshida MI, Oliveira MA, Gomes ECL, Mussel WN, Castro WV,
Enhancement of solubility and dissolution properties of lovastatin Soares CDV. Thermal characterization of lovastatin in pharma-
by liquisolid technique. IJRPC. 2014;4:713–22. ceutical formulations. J Therm Anal Calorim. 2011;106:657–64.
5. Patel MJ, Patel NM, Patel RB, Patel RP. Formulation and eval- 17. Rycerz L. Practical remarks concerning phase diagrams deter-
uation of self-microemulsifying drug delivery system of lovas- mination on the basis of differential scanning calorimetry mea-
tatin. Asian J Pharm Sci. 2010;5:266–75. surements. J Therm Anal Calorim. 2013;113:231–8.
6. Vidyadhara S, Ramana Reddy GV, Ramu A, Chandana P. For- 18. Sandhya P, Khatoon MS, Sirisha M. Improvement of solubility
mulation and evaluation of lovastatin fast dissolving tablets using by developing novel liquid dropped tablets of lovastatin. Int J
newer super disintegrants. J Pharm Res. 2011;4:1762–5. Pharm Res Rev. 2013;1:24–44.
7. Patel RP, Patel MM. Physicochemical characterization and dis- 19. Kaplan SA. Biopharmaceutical considerations in drug formation
solution study of solid dispersions of lovastatin with polyethylene design and evaluation. Drug Metab Rev. 1972;1:15–34.
glycol 4000 and polyvinylpyrrolidone K30 solid dispersions of 20. Karolewicz B, Gajda M, Owczarek A, Pluta J, Górniak A.
lovastatin. Pharm Dev Technol. 2007;2007(12):21–33. Physicochemical and dissolution studies of simvastatin solid
8. Patel M, Tekade A, Gattani S, Surana S. Solubility enhancement dispersions with Pluronic F127. Pharmazie. 2014;69:589–94.
of lovastatin by modified locust bean gum using solid dispersion
techniques. AAPS Pharm Sci Tech. 2008;9:1262–9.

123

You might also like