You are on page 1of 7

Patil Manoj D. et al.

/JPBMS 2010, 1 (16)

Available online at www.jpbms.info

Research Article
JPBMS
JOURNAL OF PHARMACEUTICAL AND BIOMEDICAL SCIENCES

FORMULATION AND EVALUATION OF SOLID DISPERSION OF TELMISARTAN WITH KOH AS ALKALISER


BY HOT MELT METHOD
Patil Manoj D.1* Keny Ramchandra V1., Saindane Dhananjay S2., Khade Trushali S.3,
Gavitre Bhaskar B.3, Kulkarni Vivek S.3, Bobe Kisan R3 and Gaikwad Dinanath T3.
1.Goa College of Pharmacy, Goa.,India
2.Gangamai College of Pharmacy, Nagoan, Dhule.,India
3.Indira Institute of Pharmacy, Sadavali(Devrukh), Dist-Ratnagiri,India

Abstract
The effect of molecular weight of polyethylene glycols (PEGS) and drug/PEG ratio on the structure and dissolution rates
of the solid dispersions with Telmisartan have been examined. Telmisartan (TEL) was chosen as a model drug due to its
poor and pH dependent water solubility. The alkalizer used to modify the pH of TEL was KOH, in the solid dispersion(SD)
system significantly increased the drug dissolution rate in gastric fluid (pH 1.2) . Structural change in drug crystallinity to
an amorphous form was also a contributing factor based on differential scanning calorimetry (DSC) thermograms and
powder X-ray diffraction (PXRD) patterns. The drug frequency of the C=O band decreased and the O–H broad band in the
Fourier transform infrared (FTIR) spectra disappeared when these alkalizer was added. It was evident that the alkalizer
in PEG 6000 based SDs synergistically enhanced dissolution of TEL not only by modulating pH but also by changing drug
crystallinity to an amorphous form via molecular interactions.

Key words: Telmisartan, Solid dispersion, Alkalizer.

Introduction
In the recent years the progress in treatment of diseases -ed molecules of which the motions are kinetically frozen.
has been evident with an upsurge in development of new A little provocation by different stressors can accelerate
drugs. An estimated 40% of these drugs are poorly water the mobility of molecules and facilitate the achievement of
soluble. Although most of the drugs have encouraging most stable and ordered crystalline states
experimental data obtained in vitro, in vivo results have Crystalline and amorphous states of a solid exhibit altered
been disappointing. The enhancement of oral molecular interactions, which may pose as energy barrier
bioavailability of poor water soluble drugs remains one of for phase transformations between them. The importance
the most challenging aspects of drug development A of pH control on the dissolution behaviors of water-
common method used to improve the dissolution rate of a insoluble drugs in SD systems became apparent a decade
poorly water soluble drug is by formation of a solid ago[4-5].
dispersion (SD) with hydrophilic polymers such as Improving drug bioavailability by changing their water
polyethylene glycol, hydroxypropylcellulose, solubility has been possible by chemical or formulation
polyvinylpyrrolidone, and other diverse carriers[1-3]. approaches Chemical approaches to improving
Various approaches have been used to improve the bioavailability without changing the active target can be
solubility of drug which in turn leads to increase in in- achieved by salt formation or by incorporating polar or
vitro drug release. The approaches are preparation of ionizable groups in the main drug structure, resulting in
amorphous form, prodrug, solid dispersion and the formation of a pro-drug. Alternatively, it has also been
complexation with various natural and synthetic reported that the modulation of pH in dosage forms is a
cyclodextrins. Out of which preparation of amorphous promising way to modify the release rate of several pH-
form is the convenient way to enhance aqueous solubility dependent and ionizable drugs[6,7]. Solid dispersions
and dissolution profile of poorly water soluble drugs. appear to be a better approach to improve drug solubility
Different techniques used to convert crystalline form of a than these techniques, because they are easier to produce
drug to the amorphous form are spray drying, jet milling, and more applicable. For instance, salt formation can only
hot melt extrusion or drug-polymer co-precipitation. be used for weakly acidic or basic drugs and not for
Materials in the amorphous form are less neutral. Furthermore, it is common that salt formation
thermodynamically stable than any crystalline form, does not achieve better bioavailability because of its in
leading to a tendency for amorphous materials to vivo conversion into acidic or basic forms. Moreover, these
transform to a known or potentially unknown crystalline types of approaches have the major disadvantage that the
form. The time scale for any transformation is also sponsoring company is obliged to perform clinical trials on
unknown and is evaluated through stability studies. these forms, since the product represents a NCE.
Amorphous state is characterized by the randomly arrang- Formulation approaches include solubilization and
particle size reduction techniques, and solid dispersions,
*Corresponding Author: among others. Solid dispersions are more acceptable to
Patil Manoj D. patients than solubilization products, since they give rise
Goa College of Pharmacy, Goa, India . to solid oral dosage forms instead of liquid as
Contact no-09970849937
1 Journal of Pharmaceutical and Biomedical Sciences (JPBMS), Vol. 01, Issue 01
Patil Manoj D. et al. /JPBMS 2010, 1 (16)

solubilization products usually do. Milling or T7 10 - 400


micronizations for particle size reduction are commonly T8 10 1 400
performed as approaches to improve solubility, on the
basis of the increase in surface area. Solid dispersions are T9 10 - 600
more efficient than these particle size reduction T10 10 - 600
techniques, since the latter have a particle size reduction
T11 10 - 600
limit around 2–5 mm which frequently is not enough to
improve considerably the drug solubility or drug release T12 10 1 600
in the small intestine and, consequently, to improve the
bioavailability . Moreover, solid powders with such a low
Fourier transforms infrared spectroscopy (FTIR)
particle size have poor mechanical properties, such as low
The spectra of the samples (again including the raw
flow and high adhesion, and are extremely difficult to
material of TEL, PEG 6000 and different SD powders) were
handle.
recorded (figure 1-3) using a spectrophotometer FTIR-
Different antihypertensive agents have been widely used
88101A (Shimadzu). KBr pellets were prepared by gently
for the treatment of hypertension. Angiotensin II receptor
mixing 1 mg of the sample with 200 mg KBr. The
antagonist are also used for the antihypertensive drugs.
wavelength ranged from 400 to 4000 cm−1 with a
Most of the Angiotensin II receptor antagonist are
resolution of 4 cm−1[12].
practically insoluble in water and thus were shown to
have dissolution rate limited oral bioavailability. Thermal analysis (DSC)
Telmisartan is a Nonpeptide Angiotensin II receptor (type A TA Instruments differential scanning calorimeter DSC-
AT1) antagonist, which is used in the treatment of 60 (shimadzu) was used to investigate the thermal
osteoarthritis, rheumatoid arthritis, acute gouty arthritis behaviors (Figure 4-6) of the raw material of TEL, PEG
and allied inflammatory conditions. TEL is manufactured 6000 and the different SD powders. The amount of sample
and supplied in the free acid form and is characterized by used ranged from 2-8mg for the SD powder and PEG 6000,
a very poor solubility, resulting in low bioavailability[8]. and was 0.4 mg for pure TEL. The samples were weighed
Telmisartan shows low aqueous solubility and poor in a standard open aluminum pan, while an empty pan of
dissolution of this molecule delays its rate of absorption the same type was used as a reference. The heat running
and finally the onset of action. Therapy with this drug for each sample was set from 30 to 300 °C at 10 °C/min,
offers a good quality of life for hypertensive patients due using nitrogen as a purge gas at heat flow of 40ml/min.[13-
14].
to the minimal side effects[9].
Powder X-ray diffraction (PXRD)
Material and Method
PXRD patterns were obtained (figure 7) with using XRD-
Materials COMPACT 3K using Powder X-ray diffractometer, CuKα
TEL was obtained as a gift sample from Ratiopharm radiation, a voltage of 40 KV and a current of 20 mA. The
Private. Ltd. (Goa, India). PEG 6000 was obtained as gift samples were scanned at the scanning rate of 0.02° /min
sample from Colorcon Asia Pvt. Ltd. (Goa, India), over the 5-60° 2θ range[15].
Potassium Hydroxide (KOH), was obtained from Loba
Scanning Electron Microscopy (SEM) analysis
Chem. (Mumbai, India). All other chemicals and reagents
The scanning electron microscope is used to study the
were of analytical grade. surface morphology of the samples. Photomicrographs of
Preparation of Solid dispersion the sample were taken (figure 8) using the Jeol Model JMS-
Solid dispersions of Telmisartan and PEG were obtained 6300 scanning electron microscope (Jeol Technics
by the hot melt method. Telmisartan: Alkalizer: PEG Company, Japan). The samples for SEM were mounted on
mixtures containing 10:1:200, 10:1:400, 10:1:600 ratios sample stubs with double sided adhesive tape, vacuum
(Table 1) were heated to 100°C with constant stirring. The coated with gold and photomicrographed at suitable
Telmisartan was miscible in the PEG melt in all magnification. The SEM uses a beam of electron to scan the
proportions with alkalizer. The melts were allowed to cool surface of the sample to build a three dimensional image
and solidify at room temperature and then stored at 4°C. of the specimen[16].
The solid product was ground in a mortar at room In-vitro dissolution
temperature and then sieved (105-250 µm). For Dissolution studies were conducted using a USP II paddle
dissolution assays, a sufficient amount of molten material method (50 rpm, 37 °C, and 900 mL dissolution medium)
(equivalent to 100 mg of Telmisartan) was poured into with a dissolution apparatus (Labinda, India). The SD
semi permeable bag (2 cm internal diameter)[10,11] powder equivalent to 80 mg TEL was exposed for 1 hr to
Table 1 : Formulation of Telmisartan Solid Dispersions gastric fluid (pH 1.2). Samples were withdrawn (Table 2)
from the dissolution medium at predetermined intervals
(10, 20, 30, 40, 50, 60, 70, 80 and 90 min) and then drug
Batch code Ratio
concentration was determined by UV (Figure 9).
Telmisartan: KOH PEG 6000 Figure 9: Dissolution profiles of T1 to T12 SD batches
T1 10 - 200 120
T1

T2 10 - 200 100 T2
T3
Drug release (%)

80
T4
T3 10 - 200 60
T5
T6

T4 10 1 200 40 T7
T8
20 T9
T5 10 - 400 T10
0 T11
0 20 40 60 80 100
T6 10 - 400 time(min)
T12

2 Journal of Pharmaceutical and Biomedical Sciences (JPBMS), Vol. 01, Issue 01


Patil Manoj D. et al. /JPBMS 2010, 1 (16)

An equivalent amount of fresh medium was added to Figure 3 : IR spectra of Telmisartan with KOH
maintain a constant dissolution volume[17].
Stability Conditions
The batches were subjected to room temperature for a
period of three months. The microcrystals were then
analyzed for the drug release profile (Table 3) by carrying
out in-vitro dissolution studies[18].
Result and Discussion
Fourier Transform Infrared Spectroscopy (FTIR)
Structural changes and the lack of a crystal structure can
lead to changes in bonding between functional groups that
can be detected by FTIR spectroscopy. Fig 1-3 shows the
FTIR spectra of Telmisartan, PEG 6000 and their SDs with
or without alkalizer. The spectrum of pure Telmisartan
showed a distinct absorption band for the carbonyl group
C=O at 1700 cm−1 and the O–H band at 3100 cm−1. Based
on the variation of these two FTIR bands, we could classify
alkalizer of the SDs into two groups. The spectra of the
KOH, showed a change in the C=O bond and the O–H bond, It has been described that a lowering of the frequency of
such that the frequency of C=O was shifted from 1695 the carbonyl stretching band from carboxylic acid is
cm−1 to 1580 cm−1 and the broad O–H band could hardly typically indicative of strong hydrogen bonding
be recognized. interactions. Moreover, the disappearance of the O–H peak
Figure 1 : IR spectra of pure Telmisartan of carboxylic acid attributable to hydrogen bonding
confirmed that this moiety could be protonated by
alkalizer mainly via a Lewis acid-base interaction .
Therefore, it was evident from FTIR spectra that there was
a molecular interaction between Telmisartan and the
alkalizer, resulting in enhanced dissolution of Telmisartan
in SDs[12].
Powder X-Ray Diffraction Studies (PXRD)
The PXRD patterns of Telmisartan, PEG 6000 and their SDs
with or without alkalizer are given in Fig. 7.
Figure 7: X-RD of Pure drug ,PEG 6000 and solid dispersion
batches

Figure 2 : IR spectra of pure Telmisartan with PEG 6000

3 Journal of Pharmaceutical and Biomedical Sciences (JPBMS), Vol. 01, Issue 01


Patil Manoj D. et al. /JPBMS 2010, 1 (16)

Figure 5 : DSC thermogram of KOH

The PXRD pattern of PEG 6000 had two characteristic


peaks of high intensity at 19.0° and 22.0°. The diffraction
pattern of pure Telmisartan was highly crystalline in
nature as indicated by numerous peaks. Three peaks at
7.0°, 14.0°, 20.0°, 23.0° and 25.0° were noticeable and the
main peak at 7.0° was particularly distinctive. It is known
that the lack of a distinctive peak of a drug in SD systems
demonstrates that a high concentration of the drug is
dissolved in the solid state. Moreover, a large reduction in Figure 6: DSC thermogram of PEG6000
characteristic peaks indicates an amorphous state. Based
on the diffractograms of SDs,we could classify the
diffraction patterns into two groups according to the
presence or absence of a Telmisartan crystalline peak at
7.0° KOH were in the group for which there was no
distinct Telmisartan crystalline peak at 7.0°.
The PXRD patterns of binary SD (Telmisartan and
PEG6000) also showed the distinctive peak at 19.0° and
22.0°, indicating that Telmisartan was still in the
crystalline form. The results of the PXRD patterns were
quite different from that of DSC thermograms In fact; PEG
6000 can play a role as the SD carrier and change the
crystalline structure of Telmisartan into a partially
crystalline structure. The incorporation of a suitable
alkalizer in PEG based SDs can definitely promote the
change of a crystalline drug to a totally amorphous form
leading to an enhanced dissolution rate[19].

Differential Scanning Calorimetry (DSC)


The DSC thermograms of TEL, PEG 6000 and their SDs are
shown in Fig. 4-6.
Figure 4 : DSC thermogrames of Pure Telmisartan
The DSC curve of pure TEL and PEG 6000 exhibited single
endothermic peaks at 269.06 °C (TEL) and 60.39 °C (PEG
6000), respectively, which corresponded to their intrinsic
melting points. The characteristic peaks of PEG 6000were
invariably identified in the DSC curves of SDs, suggesting
that PEG was present in the same physical state after
making the SD powder by the Hot Melt Method. No
characteristic melting peak of TEL was identified in the
DSC curves obtained from these PEG 6000 based SD
formulations[20].
Scanning Electron Microscopy (SEM)
Scanning electron microscopy is very helpful in studying
the change in the surface topography and shape of the
particles of pure drug and solid dispersions. The batches
of engineered crystals covering extreme polymer
concentrations from the entire range of experimental
batches were taken for SEM studies. Photomicrographs of
the pure drug (a) and SD (b) are shown in Fig 8.
Photomicrographs of the pure drug and the formulated
batches reveled the change in particle shape and surface
topography.
4 Journal of Pharmaceutical and Biomedical Sciences (JPBMS), Vol. 01, Issue 01
Patil Manoj D. et al. /JPBMS 2010, 1 (16)

Figure 8 : SEM Photomicrograph of Pure Drug (a) and SD (b)

SEM thus indicates that the polymer has formed a In -Vitro Dissolution
uniform coating over the individual drug particles thus The percentage drug release of all solid dispersion batches
resulting in the formation of spherical particles with are shown in Table 2.
improved crystal properties as revealed from later
studies.

Table 2: Cumulative % drug released of T1 to T12 SD batches

Time
( Min) T1 T2 T3 T4
0 0 0 0 0
.
10 42.9 ±2.12 34.72±2.66 17.18±2.46 51.1±1.33
20 62.65±1.26 48.16±1.33 24.31±2.68 60±2.18
30 76.53±1.78 61.02±1.56 36±2.58 72.56±2.15
40 91.19±2.77 70.13±1.89 50.44±2.90 83.3±2.43
50 106.18±2.56 80.64±2.66 59.92±1.43 94.92±2.38
60 - 87.08±2.89 72.74±2.44 -
70 - 96.52±2.56 83.99±2.87 -
80 - - 93.76±2.37 -
90 - - 99.59±2.53 -

Time
( Min) T5 T6 T7 T8
0 0 0 0 0
10 31.2±3.1 34.72±2.19 24.12±1.6 40.05±1.38
20 46.14±2.55 47.36±1.73 41.28±1.39 54.4±1.58
30 57.44±2.17 54.56±1.38 46.16±2.47 64.63±2.41
40 68.14±2.38 65.78±2.63 62.18±2.53 78.43±2.64
50 71.43±2.12 74.61±2.31 69.68±2.90 87.25±2.83
60 77.73±2.76 86.12±2.18 81.76±2.44 103.61±2.54
70 88.29±2.43 87.47±2.17 85.19±1.43 -
80 94.93±2.82 94.71±2.94 100.14±2.47 -
90 101.28±1.66 103.2±2.46 - -
Time
( Min) T9 T10 T11 T12
0 0 0 0 0
10 30.63±2.57 35.55±1.68 36.57±1.49 39.26±2.99
20 38.8±2.51 47.27±1.83 55.56±1.33 50.28±2.32
30 52.28±2.95 58.15±1.44 71.7±2.53 69.06±1.84
40 59.73±1.52 70.8±2.16 82.07±2.41 75.11±1.94
50 74.13±1.90 78.33±2.95 87.83±2.68 86.45±1.52
60 81.2±1.72 84.55±1.48 104.94±2.59 97.97±1.86
70 97.75±1.83 99.61±1.58 - -
80 101.36±1.59 - - -
90 - - - -

5 Journal of Pharmaceutical and Biomedical Sciences (JPBMS), Vol. 01, Issue 01


Patil Manoj D. et al. /JPBMS 2010, 1 (16)

The incorporation of pH modifiers was attempted due to 100% in gastric fluid with all SDs regardless of the
the fact that the chemical structure of TEL is pH- alkalizer and even with the pure drug [22].
dependent so one can assume that pH modifiers can Stability Conditions:
modulate TEL solubility leading to an increase in the Stability of the microcrystals was then analyzed for the
dissolution rate. The effect of alkalizer on TEL release drug release profiles are shown in Table 3. So this
rate in gastric fluid (pH 1.2) shown in Fig 9. Since the indicates that formulations are stable to room
drug is highly soluble in low pH conditions, the temperature for a period of three months[18].
dissolution rate of the drug after 90 min reached almost
Table 3: Stability Conditions

Time in minutes Cumulative % drug released

T1 T2 T3 T4

10
40.9 33.68 16.18 50.1
20
61.24 46.10 23.41 59.69
30
72.52 60.49 35.68 72.49
40
90.12 68.14 50.42 82.39
50
102.24 79.69 58.72 93.22
60 - -
86.08 71.54
70 - -
95.27 82.91
80 - -
92.75
90 - -
98.49

Conclusion
A significant enhancement in the aqueous solubility and the drug.DSC studies revealed that the lack of melting
dissolution profile of the drug is seen. Among the various point of solid dispersion indicated that the drug was
batches containing alkaliser being used in the study present in an amorphous form.Stability studies reveal that
namely, KOH., PEG6000 exhibited significant enhancement the product does not undergo degradation on storage and
in solubility and dissolution profile of the drug. A hence expected to maintain its integrity during storage
significant enhancement in solubility and dissolution rate with reasonable shelf life. Despite the wide application of
was seen when polymers were used in the ratio of 1:400 SDs,an obstacle of the SD method is its limited
(KOH:PEG6000) (T8).SEM studies revealed the spherical solubilization capacity. SDs containing alkalizer could be a
nature of the solid dispersion . Spherical nature indicates useful method to increase the dissolution rate of an
improvement in micromeritic properties thus anticipating ionizable drug like TEL in a pH-dependent manner. KOH in
improved tabeletting properties. Infra red study revealed the SD system significantly increased the drug dissolution
the compatibility of the drug with the polymer. XRD rate in gastric fluid (pH1.2).
diffractogram of the solid dispersions revealed a decrease
in the crystallinity of the drug reflecting amorphization of

References in polyethylene glycol 6000 solid dispersion on enhanced


1.M.-Y. Heo, Z.-Z. Piao, T.-W. Kim, Q.-R. Cao, A. Kim, B.-J. dissolution and bioavailability of ketoconazole, Arch.
Lee, Effect of solubilizing and microemulsifying excipients Pharm. Res. 28,(5),(2005),604–611.
2.N. Ahuja, P.O. Katare, B. Singh, Studies on dissolution 6.S. Li, S.Wong, S. Sethia, H. Almoazen, Y.M. Joshi, A.T.M.
enhancement and mathematical modeling of drug release Serajuddin, Investigation of solubility and dissolution of a
of a poorly water-soluble drug using water-soluble free base and two different salt forms as a function of pH,
carriers, Eur. J. Pharm. Biopharm. 65 (1) (2007) 26–38. Pharm. Res. 22 (4) (2005) 628–635.
3.T. Vasconcelos, B. Sarmento, P. Costa, Solid dispersions 7.S. Siepe, B. Lueckel, A. Krammer, A. Ries, R. Gurny,
as strategy to improve oral bioavailability of poorlywater Strategies for the design of hydrophilic matrix tablets with
soluble drugs, Drug Discov. Today 12 (23–24) (2007) controlled microenvironmental pH, Int. J. Pharm.316-1)
1068–1074. (2006) 14–20.
4.C. Doherty, P. York, Microenvironmental pH control of 8.M. Nakatani, S. Takeshi, T. Ohki, Toyoshima, Solid
drug dissolution, Int. J.Pharm. 50 (1989) 223–232. telmisartan pharmaceuticalformulations, US Patent
5.F. Usui, K. Maeda, A. Kusai, M. Ikeda, K. Nishimura, K. 2004/0110813 A1 (2004).
Yamamoto, Dissolutionimprovement of RS-8359 by the 9.M.T. Velasquez, Angiotensin II receptor blockers, a new
solid dispersion prepared by the solvent method,Int. J. class of antihypertensive drugs, Arch. Fam. Med. 5 (6)
Pharm. 170 (2) (1998) 247–256. (1996) 351–356.

6 Journal of Pharmaceutical and Biomedical Sciences (JPBMS), Vol. 01, Issue 01


Patil Manoj D. et al. /JPBMS 2010, 1 (16)

10.S. Anguiano-Igea , F. J. Otero-Espinar, J. L. Vila-Jato, J. 17.Susumu Hasegawa, Takeshi Hamauraa, Naho


Blanco-Mhdez, The properties of solid dispersions of Furuyama, Akira Kusai, Etsuo Yonemochi b, Katsuhide
clofibrate in polyethylene glycols, Pharmaceutics Acta Terada, Effects of water content in physical mixture and
Helvetiae 70 (1995) 57-66. heating temperature on crystallinity of troglitazone-PVP
11.G.R. Lloyd, D.Q.M. Craig , A. Smith , An investigation into K30 solid dispersions prepared by closed melting method,
the production of paracetamol solid dispersions in PEG Int. J. of Pharm.,302 (2005) 103–112.
4000 using hot stage differential interference contrast 18.Ghebremeskel, A.N., Vemavarapu, C., Lodaya, M., 2006.
microscopy, Int. J. of Pharm. 158 (1997) 39-46. Use of surfactants as plasticizers in preparing solid
12.Yalcın zkan, Nazan Doganay, Necati Dikmen, As¸kın dispersions of poorly soluble API: stability testing of
Isımer, Enhanced release of solid dispersions of etodolac selected solid dispersions. Pharm. Res. 23, 1928–1936.
in polyethylene Glycol, IL Farmaco 55 (2000) 433–438. 19.Adamo Finia, Jose R. Moyanob, Juan M. Ginesb, Jose I.
13.Jaymin C. Shah, Jivn R. Chen, Diana Chow, Perez-Martinezb, Antonio M. Rabasco, Diclofenac salts, II.
Preformulation study of etoposide: II. Increased solubility Solid dispersions in PEG6000 and Gelucire 50/13, Eur. J.
and dissolution rate by solid-solid dispersions, Int. J. of Pharm.and Biopharm. 60 (2005) 99–111.
Pharm. 113 (1995) 103-111. 20.Forster, J. Hempenstall, I. Tucker, T. Rades, Selection of
14.Dimitrios Bikiaris, George Z. Papageorgiou, Anagnostis excipients for melt extrusion with two poorly water-
Stergiou, Eleni Pavlidou, Evangelos Karavas, Ferras soluble drugs by solubility parameter calculation and
Kanaze, Manolis Georgarakis, Physicochemical studies on thermal analysis, Int. J. of Pharm., 226 (2001) 147–161.
solid dispersions of poorly water-soluble drugs Evaluation 21.Fude Cuia, Mingshi Yanga, Yanyan Jianga, Dongmei
of capabilities and limitations of thermal analysis Cuna, Wenhui Lina, Yuling Fan, Yoshiaki Kawashima,
techniques, Thermochimica Acta 439 (2005) 58–67. Design of sustained-release nitrendipine microspheres
15.R. Jachowicz, E. Nurnberg, B. Pieszczek, B. having solid dispersion structure by quasi-emulsion
Kluczykowska, Maciejewska, Solid dispersion of solvent diffusion method, J. Contr. Rel., 91 (2003) 375–
ketoprofen in pellets, Int. J. of Pharm., 206 (2000) 13–21. 384.
16.Hirofumi Takeuchi, Shinsuke Nagira, Hiromitsu 22.Susana Torrado, Santiago Torrado, Juan Jos Torrado,
Yamamoto, Yoshiaki Kawashima, Solid dispersion particles Rafael Caddrniga, Preparation, dissolution and
of amorphous indomethacin with fine porous silica characterization of albendazole solid dispersions, Int. J. of
particles by using spraydrying method, Int. J. of Pharm., Pharm., 140 (1996) 247-250.
293 (2005) 155–164.

7 Journal of Pharmaceutical and Biomedical Sciences (JPBMS), Vol. 01, Issue 01

You might also like