SlideShare ist ein Scribd-Unternehmen logo
1 von 6
Downloaden Sie, um offline zu lesen
 
Kharia et al: Formulation and Evaluation of Polymeric Nanoparticles of an Antiviral Drug for Gastroretention

1557  

International Journal of Pharmaceutical Sciences and Nanotechnology

Volume 4 •  Issue 4 • January – March 2012

Research Paper

MS ID: IJPSN-8-19-11-KHARIA

Formulation and Evaluation of Polymeric Nanoparticles of
an Antiviral Drug for Gastroretention
1,2

3

1

A.A. Kharia , A.K. Singhai and R. Verma
1

Oriental College of Pharmacy, Bhopal, Madhya Pradesh, India

2

Uttarakhand Technical University, Dehradun, Uttarakhand, India

3

Lakshmi Narayan College of Pharmacy, Bhopal, Madhya Pradesh, India

Received August 19, 2011; accepted January 12, 2012
ABSTRACT
The aim of present study was to formulate and evaluate
nanoparticles of acyclovir by using different hydrophilic
polymers. Acyclovir was selected as a suitable drug for gastroretentive nanoparticles due to its short half life, low
bioavailability, high frequency of administration, and narrow
absorption window in stomach and upper part of GIT. The
nano-precipitation method was used to prepare nanoparticles
so as to avoid both chlorinated solvents and surfactants to
prevent their toxic effect on the body. Nanoparticles of
acyclovir were prepared by using hydrophilic polymers such as
bovine serum albumin, chitosan, and gelatin. The prepared
formulations were then characterized for particle size,
polydispersity index, zeta potential, loading efficiency,

encapsulation efficiency and drug-excipient compatibility. The
prepared nanoparticulate formulations of acyclovir with
different polymers in 1:1 ratio have shown particle size in the
range of 250.12-743.07 nm, polydispersity index (PDI) in the
range of 0.681-1.0, zeta potential in the range of -14.2 to +33.2
mV, loading efficiency in the range of 8.74-17.54%, and
entrapment efficiency in the range of 55.7%-74.2%.
Nanoparticulate formulation prepared with chitosan in 1:1 ratio
showed satisfactory results i.e. average particle size 312.04
nm, polydispersity index 0.681, zeta potential 33.2 mV, loading
efficiency 17.54%, and entrapment efficiency 73.4%. FTIR
study concluded that no major interaction occurred between
the drug and polymers used in the present study.

KEYWORDS: Nanoparticles; gastro-retentive; nano-precipitation, polydispersity index, zeta potential; entrapment efficiency.

Introduction
The oral route of drug administration is the most
convenient and commonly used method of drug delivery
due to their considerable therapeutic advantages such as
ease of administration, patient compliance, and
flexibility in formulation (Garg et al., 2008; Gupta et al.,
2007). However, this route has several physiological
problems, such as inability to restrain and locate the
controlled drug delivery system within the desired region
of the gastrointestinal tract due to variable gastric
emptying and motility. Furthermore, the relatively brief
gastric emptying time in humans, which normally means
2-3 hours through the major absorption zone, i.e.,
stomach and upper part of the intestine, can result in
incomplete drug release from the drug delivery system
leading to reduced efficacy of the administered dose
(Rouge et al., 1996). These difficulties have prompted
researchers to design a drug delivery system which can
stay in the stomach for prolonged and predictable period
(Deshpande et al., 1996; Hwang et al., 1998). Several
attempts are being made to develop a controlled drug
delivery system, which can provide therapeutically
effective plasma drug concentration for a longer period,

thereby reducing the dosing frequency and minimizing
fluctuations in plasma drug concentration at steady-state
by delivering the drug in a controlled and reproducible
manner (Sood et al., 2003).
Different methodologies have been reported in the
literature to increase the gastric retention of drugs, like
intra-gastric floating systems, hydro dynamically
balanced
systems,
extendable
or
expandable,
microporous
compartment
system,
microballons,
bio/muco-adhesive systems, high-density systems, and
super porous biodegradable hydro gel systems (Singh
et al., 2000). After oral administration, such a dosage
form would be retained in the stomach for several hours
and would release the drug there in a controlled and
prolonged manner, so that the drug could be supplied
continuously to its absorption sites in the upper
gastrointestinal tract (Streubel et al., 2006). Prolonged
gastric retention improves bioavailability, reduces drug
waste, and improves solubility of drugs that are less
soluble in a high pH environment. It is also suitable for
local drug delivery to the stomach and proximal small
intestine (Rao et al., 2005). Gastroretention helps to
provide better availability of new products with suitable
therapeutic activity and substantial benefits for patients.

ABBREVIATIONS: Bovine serum albumin (BSA); molar (M); ultraviolet (UV); Fourier transform infra red (FTIR); polydispersity index (PDI).

1557
1558
Acyclovir, a cyclic analogue of the natural nucleoside
2-deoxyguanosine, is clinically used in the treatment of
herpes simplex, varicella zoster, cytomegalovirus, and
Epstein Barr virus infections (Brien et al., 1989).
Absorption of orally administered acyclovir is slow,
variable, and incomplete, with a bioavailability of 15%30% (Shao et al., 1994) and the elimination half-life of
acyclovir is approximately 3 hours. It has narrow
absorption window and is primarily absorbed from
stomach and upper part of the small intestine (Groning
et al., 1996); there is a need to develop an effective
formulation with enhanced gastric residence time.
Attia et al., prepared niosomes of acyclovir by using
cholesterol, span 60, and dicetyl phosphate to improve its
poor and variable bioavailability. The niosomal
dispersion when compared to the free solution has shown
more than 2-fold increase in drug bioavailability and
increase in the mean residence time of acyclovir
reflecting sustained release characteristics (Attia et al.,
2007). Palmberger et al., developed a novel oral delivery
system for the acyclovir, utilizing thiolated chitosan as
excipient, which is capable of inhibiting P-glycoprotein
(P-gp). Three chitosan-4-thiobutylamidine (Chito–TBA)
conjugates with increasing molecular mass (Chito-9.4
kDa-TBA, Chito-150 kDa-TBA, and Chito-600 kDa-TBA)
were synthesized and permeation studies on rat
intestinal mucosa and Caco-2 monolayers were
performed. They found that Chito-150 kDa–TBA/GSH
might be an appropriate sustained release drug delivery
system for acyclovir, which is able to enhance acyclovir
transport due to efflux pump inhibition (Palmberger et
al., 2008). Kharia et al., optimized the floating drug
delivery systems of acyclovir using psyllium husk and
hydroxypropylmethylcellulose K4M as the polymers by 3²
full factorial design to improve the oral bioavailability of
acyclovir. The optimized formulations followed Higuchi’s
kinetics while the drug release mechanism was found to
be anomalous type, controlled by diffusion through the
swollen matrix (Kharia et al., 2010). Stulzer et al.,
prepared microparticles containing acyclovir and
chitosan cross-linked with tripolyphosphate using the
spray-drying technique. The results obtained indicated
that the polymer/acyclovir ratio influenced the final
properties of the microparticles, with higher ratios giving
the best encapsulation efficiency, dissolution profiles,
and stability (Stulzer et al., 2009).
The aim of the present study was to formulate
gastroretentive nanoparticles of acyclovir to deliver the
drug at a controlled rate to its absorption site so that its
oral bioavailability can be enhanced. Mucoadhesive
polymers, such as bovine serum albumin, chitosan, and
gelatin, were selected to prepare gastroretentive
nanoparticles as they intensify the contact between
dosage form and the site of absorption, thereby reducing
the luminal diffusion pathway of the drug (bioadhesion)
and lead to significant improvements in oral drug
delivery (Lueben et al., 1994, Park and Robinson, 1984).
These mucoadhesive polymeric nanoparticles in the
stomach will offer various advantages such as (i) Longer
residence time of the dosage form on mucosal tissues in

Int J Pharm Sci Nanotech

Vol 4; Issue 4 • January−March 2012

the stomach. This will improve absorption of the drug
and increase the drug bioavailability. (ii) Higher drug
concentration at the site of adhesion absorption, which
will create a driving force for the paracellular passive
uptake. (iii) Immediate absorption from the bioadhesive
drug delivery system without previous dilution and
possible degradation in the luminal fluids (Hejazi and
Amiji et al., 2003)

Materials and Method
Acyclovir, bovine serum albumin (BSA), and gelatin
were obtained as a gift sample from Modern Laboratories
(Indore, India); chitosan was obtained as a gift sample
from Indian Sea Foods (Cochin, India). All other
chemicals and reagents were of laboratory grade and
were used as procured.

Preparation of Nanoparticles
Nanoparticles were prepared according to the
nanoprecipitation method with slight modification
(Elshafeey et al., 2010). Briefly, 200 mg of polymer
(bovine serum albumin, chitosan, and gelatin) was
dissolved in 25 ml of acetone separately. The acyclovir
100 mg was dissolved in 2 ml of dimethyl sulfoxide. Both
solutions were mixed and then 50 ml of water was added
and stirred for a half hour. Acetone was eliminated by
evaporation under reduced pressure using rotary flash
evaporator and the final volume of the suspension was
adjusted to 10 ml. Then this suspension was centrifuged
at 15000 rpm at 4oC for half an hour. The supernatant
was discarded and precipitate was washed 3 times with
distilled water. The nanoparticles thus obtained were
dried overnight in oven at 60ºC and stored in a
desiccator.
The prepared formulations were characterized for
loading efficiency, entrapment efficiency, particle size,
particle size distribution, polydispersity index, zeta
potential and drug excipient compatibility studies.

Characterization of Acyclovir Loaded
Nanoparticles
Loading Efficiency
Drug content in the preparation was determined by
extracting the drug from the nanoparticles with 0.1 M
hydrochloric acid. In this method, the nanoparticles (50
mg) were stirred in 50 ml of 0.1 M hydrochloric acid until
dissolved; it was filtered through a Millipore filter and
the drug content was determined, after suitable dilution,
at 254 nm by UV spectrophotometry. The loading
efficiency (L) of the nanoparticles was calculated
according to Equation 1
L (%) = (Qn /Wn) × 100
…..(1)
Where Wn is the weight of the nanoparticles and Qn
is the amount of drug present in the nanoparticles (Patel
and Patel, 2007).
1559  

Kharia et al: Formulation and Evaluation of Polymeric Nanoparticles of an Antiviral Drug for Gastroretention

Drug-loading and entrapment efficiency

Entrapment Efficiency
For determination of drug entrapment, the amount of
drug present in the clear supernatant after
centrifugation
was
determined
(w)
by
UV
spectrophotometer at 254 nm. A standard calibration
curve of drug was plotted for this purpose. The amount of
drug in supernatant was then subtracted from the total
amount of drug added during the preparation (W).
Effectively, (W-w) will give the amount of drug entrapped
in the particles (Bellare et al., 2005).
Then percentage entrapment of a drug was calculated
according to Equation 2
% Drug Entrapment = (W-w/W) × 100
…..(2)
Particle Size, Particle Size Distribution, and
Zeta Potential
The particle size and particle size distribution of the
formulation was determined by photo correlation
spectroscopy with a zeta master (Malvern Instruments,
UK) equipped with the Malvern PCS software. Every
sample was diluted with distilled water. The surface
charge (Zeta potential) was determined by measuring the
electrophoretic mobility of the nanoparticles using a
Malvern zeta sizer (Malvern Instruments, UK). Samples
were prepared by diluting with distilled water
(Pignatello et al., 2006).
Polydispersity Index
Polydispersity index is a parameter to define the
particle size distribution of nanoparticles obtained from
photon correlation spectroscopic analysis. It is a
dimensionless
number
extrapolated
from
the
autocorrelation function and ranges from a value of 0.01
for mono dispersed particles and up to values of 0.5-0.7.
Samples with very broad size distribution have
polydispersity index values > 0.7 (Nidhin et al., 2008).
Drug-Excipient Compatibility Studies
The drug excipient compatibility studies was
performed by using FT-IR spectrophotometer (Perkin
Elmer). The FT-IR spectra of drug, polymers, and
formulations were analyzed separately and then
correlated for incompatibility.

Results and Discussion
The method of nanoprecipitation was used so as to
avoid both chlorinated solvents and surfactants to
prevent their toxic effect on the body. All the
determinations were done in triplicate.

Although drug loading expresses the percent weight
of active ingredient encapsulated to the weight of
nanoparticles, entrapment efficiency is the ratio of the
experimentally determined percentage of drug content
compared with actual, or theoretical mass, of drug used
for the preparation of the nanoparticles. The loading
efficiency depends on the polymer-drug combination and
the method used. Hydrophobic polymers encapsulate
larger amounts of hydro phobic drugs, whereas
hydrophilic polymers entrap greater amounts of more
hydrophilic drugs. Several formulation parameters, such
as emulsifier type, weight ratio of polymer to drug, and
organic to aqueous phase ratio, will influence the extent
of drug loading.
The effect of polymer on drug loading efficiency and
entrapment efficiency are given in Table 1 and shown in
Figure 1. The values were in the range of 8.74%-17.54%
and 55.7%-74.2%, respectively. Loading efficiency was
low for gelatin and albumin nanoparticles (8.74% and
11.43%
respectively)
while
high
for
chitosan
nanoparticles (17.54%). It was found that the
entrapment efficiency were high for the formulations
containing chitosan and gelatin (73.4% and 74.2%
respectively) while low for the formulation containing
bovine serum albumin (55.7%). Loading efficiency may be
increased by increasing polymer ratio, so that sufficient
quantity of polymer will be available to entrap the drug
present in the solution, while less entrapment efficiency
may be due to hydrophilic nature of acyclovir.

Particle Size Distribution and
Polydispersity Index
The particle size and particle size distribution are
critical factors in the performance of nanoparticles, as
batches with wide particle size distribution show
significant variations in drug loading, drug release,
bioavailability, and efficacy. Particle size and particle
size distribution can be determined using light scattering
techniques and by scanning or transmission electron
microscopy. Formulation of nanoparticles with a narrow
size distribution will be a challenge if emulsion cannot be
produced with a narrow droplet size distribution. As
nanoparticles are internalized into cells by endocytosis,
an increase in particle size will decrease uptake and
potentially, affect bioavailability of the drug. The extent
of endocytosis is dependent on the type of the target cell.

TABLE 1
Drug-loading and entrapment efficiency.
Sl.
No.

Formulation
Code

Polymer

Drug:Polymer
ratio

Formulation
Code

Loading
Efficiency* ± SD %

Entrapment
Efficiency* ± SD %

1.
2.
3.

NP 1
NP 2
NP 3

Bovine Serum Albumin
Chitosan
Gelatin

1:1
1:1
1:1

NP 1
NP 2
NP 3

11.43±2
17.54±3
8.74±3

55.7±6.3
73.4±3.5
74.2±4.9

* = Average of three determinations
1560

Int J Pharm Sci Nanotech

Vol 4; Issue 4 • January−March 2012

Fig. 1. Effect of polymer on loading
and entrapment efficiency.

The results of prepared nanoparticulate formulations
of acyclovir with different polymers are given in Table 2
and shown in Figure 2. The formulations had very high
polydispersity index (PDI) in the range of 0.681-1.0.
From the particle size distribution data, it is evident that
in case of BSA nanoparticles, mean particle diameter
was 250.12 nm and major portion of the particles were in
the range of 200-400 nm, for chitosan nanoparticles
mean particle diameter was 312.04 nm; and major
portion of the particles were in range of 200-525 nm. In
case of gelatin nanoparticles mean particle diameter was
743.07 nm and most of the particles were in the range of
480-1200 nm. However, in all the formulations contained
a minority population of nanoparticles in much smaller
range. For BSA, about 10.1% of the particles were in the
range 15-30 nm, for chitosan about 7.1% of the particles
were in the range 48-90 nm and for gelatin 14.1% of the
particles were in the range 70-160 nm. These minority
populations are responsible for larger over all
polydispersity indices of the formulations. We are
currently exploring the process variables affecting the
relative amounts of different populations with an
objective to increase the yield of the particles in the
smaller range to get much smaller nanoparticles, which
have
greater
degree of
monodispersity.
Such
nanoparticles can be easily separated from the larger
sized population by simple methods like filtration.
From the above data it is clear that nanoparticles
prepared by using chitosan and BSA exhibited reduction
in mean nanoparticulate diameter and narrower
granulometric distribution. But the nanoparticles
prepared using gelatin as a polymer resulted in
nanoparticulate population of large particles. The higher
particle size and polydispersity index may be because of
absence of emulsifier as the use of emulsifier decreases
the surface tension between organic phase acetone and
aqueous phase and leads to the formation of smaller
solvent droplets, which in turn causes decrease in
particle size. It also stabilizes newly generated surfaces

and prevents aggregation of the particles as reported by
previous researchers (Schubert and Muller, 2003).
Therefore results which were obtained in this study may
be improved by using increased drug:polymer ratio, using
different formulation strategy such as desolvation (for
gelatin and albumin) or counter ion induced aggregation
(for chitosan and sodium alginate), employing cross
linking agent followed by neutralizing residual cross
linking agent with cysteine and high speed stirring.

Zeta Potential
The measurement of the zeta potential allows
predictions about the storage stability of colloidal
dispersions. In general, particle aggregation is less likely
to occur for charged particles (i.e. high zeta potential)
due to electric repulsion. Generally, Zeta potential values
above 30 mV (positive or negative values) lead to more
stable nanocapsule suspensions because repulsion
between the particles prevented their aggregation. A
decrease in zeta potential, i.e. electrostatic repulsion,
was considered as the cause for the aggregation process
(Dalengon et al., 1998). The charge on the surface of the
nanospheres will influence their distribution in the body
and the extent of uptake into the cells. Because cell
membranes are negatively charged, there is greater
electrostatic affinity for positively charged nanoparticles.
Therefore, the surface of cationic or neutral
nanoparticles may be modified to confer a positive charge
to enhance efficacy.
The zeta potential values which were in the range of
–14.2 - +21.7 mV, indicates that the colloidal suspension
may not be stable and may lead to aggregation. Zeta
potential values can be altered by modifying the major
components such as surfactants, polymer, and surface
composition of the nanoparticles, the presence or the
absence of adsorbed compounds, composition of the
dispersing phase, mainly the ionic strength, and the pH
(Barratt et al., 1999).
1561  

Kharia et al: Formulation and Evaluation of Polymeric Nanoparticles of an Antiviral Drug for Gastroretention

during the process of formulation polymer has not
reacted with the drug to give rise to reactant products.
So it is only physical mixture and there is no interaction
between them which is in favor to proceed for
formulation.

Drug-Excipient Compatibility Studies
The results of drug-excipient compatibility studies
are shown in Table 3. From the IR data it is clear that
functionalities of drug have remained unchanged,
including intensities of the peak. This suggests that
TABLE 2

Drug polymer ratio, mean particle size, particle size distribution, poly dispersity index (PDI) and zeta potential.
Sl.
No.

Formulation
Code

Polymer

Mean Particle Size
(nm) ± SD

Size Distribution

PDI ± SD

Zeta Potential
(mV) ± SD

1.

NP 1

Bovine Serum Albumin

250.12±18

10.1% (15-30 nm)
89.9 % (200-400 nm)

1.0±0.12

21.7±1.4

2.

NP 2

Chitosan

312.04±32

7.8% (48-90 nm)
92.2% (200-525 nm)

0.681±0.15

33.2±2.1

3.

NP 3

Gelatin

743.07±45

14.2% (70-160 nm)
85.8% (480-1200 nm)

0.7770±0.14

-14.2±1.3

* = Average of three determination

Fig. 2. Effect of polymer on mean
particle size.

TABLE 3
Major peaks observed in the FT-IR spectrum.
S.
No.

1.
2.
3.
4.
5.
6.
7.
8.

Bands

-OH
N-H bend, assy.
C-C ring str.
Aromatic amines,
C-N str.
Assy. C-O-C str.
Sym C-O-C str.
N-H wagging
Mono-substitution
in ring

Drug

NP 1

cm-1

cm-1

NP 2

NP 3

cm-1

3447.8 and 3337.9
1632.5 cm-1
1537 and 1483.7 cm-1
1305.9 and 1392.1 cm-1

3442.9
1626.4 cm-1
1479.2 and 1531.1 cm-1
1387.1 cm-1

3434.5 and 3324.5
1638.8 cm-1
1456.9 cm-1
1390.4 and 1302.5 cm-1

3441.1 cm-1
1635.4 cm-1
1532.1 and 1481.1 cm-1
1303.6 cm-1

1219 cm-1
1013 cm-1
900.5 and 864.2 cm-1
777.1,753.7 and 681.2 cm-1

1216.8 cm-1
1011.9 cm-1
900.1 cm-1
767.6 and 672 cm-1

1219.2 cm-1
1080.3 cm-1
897.3 cm-1
771.5 and 675.5 cm-1

1216.5 cm-1
1011.2 cm-1
899.5 cm-1
769.8 and 670.9 cm-1

NP1 = Bovine Serum Albumin Nanoparticles, NP2 = Chitosan Nanoparticles, NP3 = Gelatin Nanoparticles

Conclusion
Among different nanoparticulate formulations
prepared by nanoprecipitation method formulation NP 2,
with chitosan in 1:1 drug: polymer ratio, showed
satisfactory results; i.e. mean particle size of 312.04 nm
(majority of the particles were in the range of 200-525

nm), polydispersity index of 0.681, zeta potential of 33.2
and loading efficiency of 17.54%, and entrapment
efficiency of 73.4%. FTIR study concluded that no major
interaction occurred between the drug and polymers used
in the present study.
1562
Acknowledgements
The authors are thankful to Modern Laboratories
(Indore, India) and Indian Sea Foods (Cochin, India) for
providing gift samples and Director, Oriental College of
Pharmacy, Bhopal, India for their support and
cooperation in carrying out the research work. The
authors are very thankful to SAIL, RGPV, Bhopal, for
their valuable contribution in the determination of
particle size, PDI and zeta potential of the formulations.
References
Attia IA, El-Giawy SA, Fouda MA and Donia AM (2007). Influence of
a niosomal formulation on the oral bioavailability of acyclovir in
rabbits. AAPS Pharm SciTech 8; 106: E1-E27.
Barratt G (1999). Characterization of colloidal drug carrier systems
with zeta potential measurements. Pharma Tech Eur 11: 25–32.
Bellare J, Banerjee R and Das S (2005). Aspirin loaded albumin
nanoparticles by coacervation: Implications in drug delivery.
Trends Bio Artif Organs 18: 203-211.
Dalengon F, Amjaud Y, Lafforgue C, Derouin F, Fessi H (1998).
Atovaquone and rifabutine-loaded nanocapsules: Formulation
studies. Int J Pharm 153: 127-130.
Deshpande AA, Rhodes TN and Shah H (1996). Controlled release
drug delivery systems for prolonged gastric residence an
overview. Drug dev Ind pharm 22: 531-539.
Elshafeey AH, Kamel AO and Awad GAS (2010). Ammonium
methacrylate unit’s polymer content and their effect on acyclovir
colloidal nanoparticles properties and bioavailability in human
volunteers. Colloids and Surf B: Biointerfaces 75: 398-404.
Garg R and Gupta GD (2008). Progress in controlled gastroretentive
delivery systems. Trop J Pharm Res 7: 1055-1066.
Groning R, Berntgen M and Geogarakis M (1996). Acyclovir serum
concentrations following peroral administration of magnetic
depot tablets and the influence of extra corporal magnet to
control gastrointestinal transit. Eur J Pharm Biopharm 46: 28591.
Gupta SN and Aggarwal N (2007). A gastro-retentive floating
delivery system for 5-Fluorouracil. Asian Journal of
Pharmaceutical Sciences 2: 143-149.
Hejazi R and Amiji M (2003). Chitosan-based gastrointestinal
delivery systems. J Control Release 89: 151-165.
Hwang SJ, Park H and Park K (1998). Gastric retentive drug
delivery systems. Crit Rev Ther Drug 15: 243-248.
Kharia AA, Hiremath SN, Singhai AK, Omray LK, Jain SK (2010).
Design and optimization of floating drug delivery system of
acyclovir. Indian J Pharm Sci 72: 599-606.
Lueben HL, Lehr CM, Rentel CO, Noach ABJ, Boer AGD, Verhoef
JC and Junginger HE (1994). Bioadhesive polymers for the
peroral delivery of peptide drugs. J Control Release 29: 329-338

Int J Pharm Sci Nanotech

Vol 4; Issue 4 • January−March 2012

Nidhin M, Indumathy R, Sreeram KJ and Balachandran U (2008).
Synthesis of iron oxide nanoparticles of narrow size distribution
on polysaccharide templates. Bull Mater Sci 31: 93-96.
O’Brien J and Campoli D (1989). Acyclovir: an updated review of its
antiviral activity, pharmacokinetics properties and therapeutic
efficacy. Drugs 37: 233-309.
Palmberger TF, Hpmbach J and Schnurch AB (2008). Thiolated
chitosan: Development and in vitro evaluation of an oral delivery
system for acyclovir. Int J Pharm 348: 54-60.
Park K and Robinson JR (1984). Bioadhesive polymers as platforms
for oral-controlled drug delivery: method to study bioadhesion.
Int J Pharm 19: 107-127.
Patel JK and Patel MM (2007). Stomach Specific Anti-Helicobacter
Pylori Therapy: Preparation and Evaluation of Amoxicillin –
Loaded Chitosan Muco adhesive Microspheres. Curr Drug Deliv
4: 41-50.
Pignatello R, Ricupero N, Bucolo, Maugeri F, Maltese A and Puglisi
G (2006). Preparation and characterization of eudragit retard
nanasuspensions for the ocular delivery of cloricromene. AAPS
Pharm SciTech 24; 7: E27.
Rao PB, Kottan NA and Snehith VS (2005). Development of
gastroretentive drug delivery system of cephalexin by using
factorial design. ARS Pharmaceutica 50: 8-24.
Rouge N, Buri P and Doelke E (1996). Drug absorption sites in the
gastrointestinal tract and dosage forms for site specific delivery.
Int J Pharm 136:117-139.
Schubert MA and Muller GCC. Solvent injection as a new approach
for manufacturing lipid nanoparticles-evaluation of the method
and process parameters. Eur J Pharm Biopharm 2003;55:125131
Shao Z and Mitra AK (1994). Bile salts-fatty acid mixed micelles as
nasal absorption promotors: III. Effect on nasal transport and
enzymatic degradation of acyclovir prodrug. Pharm Res 11: 24350.
Singh BN and Kim KH (2000). Floating drug delivery systems: an
approach to oral controlled drug delivery via gastric retention. J
Control Release 63: 235-239.
Sood A and Panchagnula R (2003). Design of controlled release
delivery systems using modified pharmacokinetic approach: a
case study for drugs having a short elimination half life and a
narrow therapeutic index. Int J Pharm 261: 27-41.
Streubel A, Siepmann J and Bodmeier R (2006). Gastroretentive
drug delivery system. Expert Opin Drug Deliv 3: 217-233.
Stulzer HK, Tagliari MP, Parize AL, Silva MAS and Laranjeira
MCM (2009). Evaluation of cross-linked chitosan microparticles
containing acyclovir obtained by spray-drying. Mater Sci Eng C
29: 387-392.
Address correspondence to: Ankit Anand Kharia, Oriental College of
Pharmacy Raisen Road, Thakral Nagar, BHOPAL (M.P.) 462021.
Mob: 09685360536; Email: ankitanandkharia@yahoo.co.in

Weitere ähnliche Inhalte

Was ist angesagt?

Gastroretentive drug delivery system
Gastroretentive drug delivery systemGastroretentive drug delivery system
Gastroretentive drug delivery systemArpitha Aarushi
 
Formulation Approaches For Gastro Retentive Drug Delivery
Formulation Approaches For Gastro Retentive Drug DeliveryFormulation Approaches For Gastro Retentive Drug Delivery
Formulation Approaches For Gastro Retentive Drug DeliveryNaman Pant
 
Design of gastroretentive bilayer floating films of propranolol hydrochloride...
Design of gastroretentive bilayer floating films of propranolol hydrochloride...Design of gastroretentive bilayer floating films of propranolol hydrochloride...
Design of gastroretentive bilayer floating films of propranolol hydrochloride...Namdeo Shinde
 
Formulation Development and Evaluation of Gastro Retentive Drug Delivery Syst...
Formulation Development and Evaluation of Gastro Retentive Drug Delivery Syst...Formulation Development and Evaluation of Gastro Retentive Drug Delivery Syst...
Formulation Development and Evaluation of Gastro Retentive Drug Delivery Syst...Dr. Raghavendra Kumar Gunda
 
evaluation of gastro retentive drug delivery system (GRDDS)
evaluation of gastro retentive drug delivery system (GRDDS)evaluation of gastro retentive drug delivery system (GRDDS)
evaluation of gastro retentive drug delivery system (GRDDS)Imran Pasha
 
Colon Specific Drug Delivery System
Colon Specific Drug Delivery SystemColon Specific Drug Delivery System
Colon Specific Drug Delivery Systemadsavaliya
 
Colon drug delivery system ppt
Colon drug delivery system pptColon drug delivery system ppt
Colon drug delivery system pptMujeeb Rehman
 
Drug Absorption Studies 2008
Drug Absorption Studies  2008Drug Absorption Studies  2008
Drug Absorption Studies 2008Sainal Edi Kamal
 
Colon drug delivery system
Colon drug delivery systemColon drug delivery system
Colon drug delivery systemForam Machhar
 
Shivaji shinde colloquium ppt..
Shivaji shinde colloquium ppt..Shivaji shinde colloquium ppt..
Shivaji shinde colloquium ppt..sushiv
 
Gestro retention drug delivery system, by dr. umesh kumar sharma & arathy...
Gestro retention drug delivery system, by dr. umesh kumar sharma & arathy...Gestro retention drug delivery system, by dr. umesh kumar sharma & arathy...
Gestro retention drug delivery system, by dr. umesh kumar sharma & arathy...Dr. UMESH KUMAR SHARMA
 
Sagar grdds final ppt pd
Sagar grdds final ppt pdSagar grdds final ppt pd
Sagar grdds final ppt pdSagar Bansal
 
Concepts of controlled and novel drug delivery system
Concepts of controlled and novel drug delivery systemConcepts of controlled and novel drug delivery system
Concepts of controlled and novel drug delivery systemPratiksha Chandragirivar
 
Colon Specific Drug Delivery System: Basics and Approaches
Colon Specific Drug Delivery System: Basics and ApproachesColon Specific Drug Delivery System: Basics and Approaches
Colon Specific Drug Delivery System: Basics and ApproachesNone
 
Biopharmaceutical classification system
Biopharmaceutical classification systemBiopharmaceutical classification system
Biopharmaceutical classification systemArpitha Aarushi
 
Colon targeted drug delivery
Colon targeted drug deliveryColon targeted drug delivery
Colon targeted drug deliveryPrachi Joshi
 
colon targetting
colon targettingcolon targetting
colon targettingROHIT
 

Was ist angesagt? (20)

Colon specific DDS
Colon specific DDSColon specific DDS
Colon specific DDS
 
Gastroretentive drug delivery system
Gastroretentive drug delivery systemGastroretentive drug delivery system
Gastroretentive drug delivery system
 
Formulation Approaches For Gastro Retentive Drug Delivery
Formulation Approaches For Gastro Retentive Drug DeliveryFormulation Approaches For Gastro Retentive Drug Delivery
Formulation Approaches For Gastro Retentive Drug Delivery
 
Design of gastroretentive bilayer floating films of propranolol hydrochloride...
Design of gastroretentive bilayer floating films of propranolol hydrochloride...Design of gastroretentive bilayer floating films of propranolol hydrochloride...
Design of gastroretentive bilayer floating films of propranolol hydrochloride...
 
Formulation Development and Evaluation of Gastro Retentive Drug Delivery Syst...
Formulation Development and Evaluation of Gastro Retentive Drug Delivery Syst...Formulation Development and Evaluation of Gastro Retentive Drug Delivery Syst...
Formulation Development and Evaluation of Gastro Retentive Drug Delivery Syst...
 
evaluation of gastro retentive drug delivery system (GRDDS)
evaluation of gastro retentive drug delivery system (GRDDS)evaluation of gastro retentive drug delivery system (GRDDS)
evaluation of gastro retentive drug delivery system (GRDDS)
 
Colon Specific Drug Delivery System
Colon Specific Drug Delivery SystemColon Specific Drug Delivery System
Colon Specific Drug Delivery System
 
Colon drug delivery system ppt
Colon drug delivery system pptColon drug delivery system ppt
Colon drug delivery system ppt
 
Drug Absorption Studies 2008
Drug Absorption Studies  2008Drug Absorption Studies  2008
Drug Absorption Studies 2008
 
Colon drug delivery system
Colon drug delivery systemColon drug delivery system
Colon drug delivery system
 
Shivaji shinde colloquium ppt..
Shivaji shinde colloquium ppt..Shivaji shinde colloquium ppt..
Shivaji shinde colloquium ppt..
 
Floating drug delivery systems
Floating drug delivery systemsFloating drug delivery systems
Floating drug delivery systems
 
Gestro retention drug delivery system, by dr. umesh kumar sharma & arathy...
Gestro retention drug delivery system, by dr. umesh kumar sharma & arathy...Gestro retention drug delivery system, by dr. umesh kumar sharma & arathy...
Gestro retention drug delivery system, by dr. umesh kumar sharma & arathy...
 
Grdds ppt
Grdds pptGrdds ppt
Grdds ppt
 
Sagar grdds final ppt pd
Sagar grdds final ppt pdSagar grdds final ppt pd
Sagar grdds final ppt pd
 
Concepts of controlled and novel drug delivery system
Concepts of controlled and novel drug delivery systemConcepts of controlled and novel drug delivery system
Concepts of controlled and novel drug delivery system
 
Colon Specific Drug Delivery System: Basics and Approaches
Colon Specific Drug Delivery System: Basics and ApproachesColon Specific Drug Delivery System: Basics and Approaches
Colon Specific Drug Delivery System: Basics and Approaches
 
Biopharmaceutical classification system
Biopharmaceutical classification systemBiopharmaceutical classification system
Biopharmaceutical classification system
 
Colon targeted drug delivery
Colon targeted drug deliveryColon targeted drug delivery
Colon targeted drug delivery
 
colon targetting
colon targettingcolon targetting
colon targetting
 

Andere mochten auch

Nanopharmaceutical & its_industrial_applications_program-ppt
Nanopharmaceutical & its_industrial_applications_program-pptNanopharmaceutical & its_industrial_applications_program-ppt
Nanopharmaceutical & its_industrial_applications_program-ppttabirsir
 
SSDN SACHIN LPU GUIDE MR. NARENDRA PANDEY
SSDN SACHIN LPU GUIDE MR. NARENDRA PANDEYSSDN SACHIN LPU GUIDE MR. NARENDRA PANDEY
SSDN SACHIN LPU GUIDE MR. NARENDRA PANDEYsachin
 
Nanoparticles based drug delivery systems for treatment of
Nanoparticles based drug delivery systems for treatment ofNanoparticles based drug delivery systems for treatment of
Nanoparticles based drug delivery systems for treatment ofaisha rauf
 
Oral Dispersible Tablets
Oral Dispersible TabletsOral Dispersible Tablets
Oral Dispersible TabletsSuresh Rewar
 
Liposomes- A Novel Drug Delivery System
Liposomes- A Novel Drug Delivery SystemLiposomes- A Novel Drug Delivery System
Liposomes- A Novel Drug Delivery SystemAPCER Life Sciences
 
Formulation and evaluation of
Formulation and evaluation ofFormulation and evaluation of
Formulation and evaluation ofGajanan Ingole
 
Formulation development and evalution of matrix tablet of
Formulation development and evalution of matrix tablet ofFormulation development and evalution of matrix tablet of
Formulation development and evalution of matrix tablet ofGajanan Ingole
 
NANOPARTICULATE DRUG DELIVERY SYSTEM
NANOPARTICULATE DRUG DELIVERY SYSTEMNANOPARTICULATE DRUG DELIVERY SYSTEM
NANOPARTICULATE DRUG DELIVERY SYSTEMSagar Savale
 
Formulation and evaluation of fast dissolving tablet- by aryan and rajesh
Formulation and evaluation of fast dissolving tablet- by aryan and rajeshFormulation and evaluation of fast dissolving tablet- by aryan and rajesh
Formulation and evaluation of fast dissolving tablet- by aryan and rajeshSridhar Sri
 
M. Pharm. Thesis Presentation
M. Pharm. Thesis PresentationM. Pharm. Thesis Presentation
M. Pharm. Thesis Presentationvibhasharma11
 
liposomes and niosomes
liposomes and niosomes liposomes and niosomes
liposomes and niosomes swethaaitha
 

Andere mochten auch (15)

Nanopharmaceutical & its_industrial_applications_program-ppt
Nanopharmaceutical & its_industrial_applications_program-pptNanopharmaceutical & its_industrial_applications_program-ppt
Nanopharmaceutical & its_industrial_applications_program-ppt
 
SSDN SACHIN LPU GUIDE MR. NARENDRA PANDEY
SSDN SACHIN LPU GUIDE MR. NARENDRA PANDEYSSDN SACHIN LPU GUIDE MR. NARENDRA PANDEY
SSDN SACHIN LPU GUIDE MR. NARENDRA PANDEY
 
Nanoparticles based drug delivery systems for treatment of
Nanoparticles based drug delivery systems for treatment ofNanoparticles based drug delivery systems for treatment of
Nanoparticles based drug delivery systems for treatment of
 
ODT project protocol-Reshma
ODT project protocol-ReshmaODT project protocol-Reshma
ODT project protocol-Reshma
 
Oral Dispersible Tablets
Oral Dispersible TabletsOral Dispersible Tablets
Oral Dispersible Tablets
 
Liposomes
LiposomesLiposomes
Liposomes
 
Liposomes- A Novel Drug Delivery System
Liposomes- A Novel Drug Delivery SystemLiposomes- A Novel Drug Delivery System
Liposomes- A Novel Drug Delivery System
 
Liposomes
LiposomesLiposomes
Liposomes
 
Formulation and evaluation of
Formulation and evaluation ofFormulation and evaluation of
Formulation and evaluation of
 
Formulation development and evalution of matrix tablet of
Formulation development and evalution of matrix tablet ofFormulation development and evalution of matrix tablet of
Formulation development and evalution of matrix tablet of
 
NANOPARTICULATE DRUG DELIVERY SYSTEM
NANOPARTICULATE DRUG DELIVERY SYSTEMNANOPARTICULATE DRUG DELIVERY SYSTEM
NANOPARTICULATE DRUG DELIVERY SYSTEM
 
Formulation and evaluation of fast dissolving tablet- by aryan and rajesh
Formulation and evaluation of fast dissolving tablet- by aryan and rajeshFormulation and evaluation of fast dissolving tablet- by aryan and rajesh
Formulation and evaluation of fast dissolving tablet- by aryan and rajesh
 
M. Pharm. Thesis Presentation
M. Pharm. Thesis PresentationM. Pharm. Thesis Presentation
M. Pharm. Thesis Presentation
 
liposomes and niosomes
liposomes and niosomes liposomes and niosomes
liposomes and niosomes
 
Liposome ppt
Liposome pptLiposome ppt
Liposome ppt
 

Ähnlich wie 1557 full

GASTRO RETENTIVE DRUG DELIVERY SYSTEM PPT PDF.pdf
GASTRO RETENTIVE DRUG DELIVERY SYSTEM PPT PDF.pdfGASTRO RETENTIVE DRUG DELIVERY SYSTEM PPT PDF.pdf
GASTRO RETENTIVE DRUG DELIVERY SYSTEM PPT PDF.pdfVIJAY JADHAV
 
FORMULATION DEVELOPMENT AND EVALUATION OF CARVEDILOL PHOSPHATE GASTRO RETENTI...
FORMULATION DEVELOPMENT AND EVALUATION OF CARVEDILOL PHOSPHATE GASTRO RETENTI...FORMULATION DEVELOPMENT AND EVALUATION OF CARVEDILOL PHOSPHATE GASTRO RETENTI...
FORMULATION DEVELOPMENT AND EVALUATION OF CARVEDILOL PHOSPHATE GASTRO RETENTI...Dr. Raghavendra Kumar Gunda
 
Protocol proposal to university
Protocol proposal to universityProtocol proposal to university
Protocol proposal to universitysushiv
 
M.Pham project presentation phase 2
M.Pham project presentation phase 2M.Pham project presentation phase 2
M.Pham project presentation phase 2Dhaneshwar P
 
Design, Formulation and Evaluation of Atenolol Gastro Retentive Floating Tablets
Design, Formulation and Evaluation of Atenolol Gastro Retentive Floating TabletsDesign, Formulation and Evaluation of Atenolol Gastro Retentive Floating Tablets
Design, Formulation and Evaluation of Atenolol Gastro Retentive Floating TabletsDr. Raghavendra Kumar Gunda
 
Floating microspheres: Research article
Floating microspheres: Research articleFloating microspheres: Research article
Floating microspheres: Research articleSriramNagarajan19
 
Formulation and Evaluation of Floating in Situ Gel of Lafutidine
Formulation and Evaluation of Floating in Situ Gel of LafutidineFormulation and Evaluation of Floating in Situ Gel of Lafutidine
Formulation and Evaluation of Floating in Situ Gel of LafutidineAI Publications
 
Formulation and Evaluation of Floating Matrix Tablets of an Antipsychotic Drug
Formulation and Evaluation of Floating Matrix Tablets of an Antipsychotic DrugFormulation and Evaluation of Floating Matrix Tablets of an Antipsychotic Drug
Formulation and Evaluation of Floating Matrix Tablets of an Antipsychotic DrugBRNSSPublicationHubI
 
Formulation and Evaluation of Gabapentin Mucoadhesive Gastro Retentive Tablets.
Formulation and Evaluation of Gabapentin Mucoadhesive Gastro Retentive Tablets.Formulation and Evaluation of Gabapentin Mucoadhesive Gastro Retentive Tablets.
Formulation and Evaluation of Gabapentin Mucoadhesive Gastro Retentive Tablets.pharmaindexing
 
Aarti grdds ppt
Aarti grdds pptAarti grdds ppt
Aarti grdds pptDVVPCOP
 
DEVELOPMENT AND OPTIMIZATION OF CIPROFLOXACIN HCL HOLLOW MICROSPHERES (MICRO ...
DEVELOPMENT AND OPTIMIZATION OF CIPROFLOXACIN HCL HOLLOW MICROSPHERES (MICRO ...DEVELOPMENT AND OPTIMIZATION OF CIPROFLOXACIN HCL HOLLOW MICROSPHERES (MICRO ...
DEVELOPMENT AND OPTIMIZATION OF CIPROFLOXACIN HCL HOLLOW MICROSPHERES (MICRO ...ANURAG GROUP OF INSTITUTIONS
 
Approaches Of Gastro-Retentive Drug Delivery System or GRDDS
Approaches Of Gastro-Retentive Drug Delivery System or GRDDSApproaches Of Gastro-Retentive Drug Delivery System or GRDDS
Approaches Of Gastro-Retentive Drug Delivery System or GRDDSAkshayPatane
 
Formulation Design, Optimization and Evaluation of Carvedilol Phosphate Gastr...
Formulation Design, Optimization and Evaluation of Carvedilol Phosphate Gastr...Formulation Design, Optimization and Evaluation of Carvedilol Phosphate Gastr...
Formulation Design, Optimization and Evaluation of Carvedilol Phosphate Gastr...Dr. Raghavendra Kumar Gunda
 

Ähnlich wie 1557 full (20)

GASTRO RETENTIVE DRUG DELIVERY SYSTEM PPT PDF.pdf
GASTRO RETENTIVE DRUG DELIVERY SYSTEM PPT PDF.pdfGASTRO RETENTIVE DRUG DELIVERY SYSTEM PPT PDF.pdf
GASTRO RETENTIVE DRUG DELIVERY SYSTEM PPT PDF.pdf
 
A Study on Formulation of Multiparticulate Gastro Retentive Drug Delivery Sys...
A Study on Formulation of Multiparticulate Gastro Retentive Drug Delivery Sys...A Study on Formulation of Multiparticulate Gastro Retentive Drug Delivery Sys...
A Study on Formulation of Multiparticulate Gastro Retentive Drug Delivery Sys...
 
FORMULATION DEVELOPMENT AND EVALUATION OF CARVEDILOL PHOSPHATE GASTRO RETENTI...
FORMULATION DEVELOPMENT AND EVALUATION OF CARVEDILOL PHOSPHATE GASTRO RETENTI...FORMULATION DEVELOPMENT AND EVALUATION OF CARVEDILOL PHOSPHATE GASTRO RETENTI...
FORMULATION DEVELOPMENT AND EVALUATION OF CARVEDILOL PHOSPHATE GASTRO RETENTI...
 
Protocol proposal to university
Protocol proposal to universityProtocol proposal to university
Protocol proposal to university
 
M.Pham project presentation phase 2
M.Pham project presentation phase 2M.Pham project presentation phase 2
M.Pham project presentation phase 2
 
04_AJP_460_15_OA_20151231_V1
04_AJP_460_15_OA_20151231_V104_AJP_460_15_OA_20151231_V1
04_AJP_460_15_OA_20151231_V1
 
Design, Formulation and Evaluation of Atenolol Gastro Retentive Floating Tablets
Design, Formulation and Evaluation of Atenolol Gastro Retentive Floating TabletsDesign, Formulation and Evaluation of Atenolol Gastro Retentive Floating Tablets
Design, Formulation and Evaluation of Atenolol Gastro Retentive Floating Tablets
 
Floating microspheres: Research article
Floating microspheres: Research articleFloating microspheres: Research article
Floating microspheres: Research article
 
Formulation and Evaluation of Floating in Situ Gel of Lafutidine
Formulation and Evaluation of Floating in Situ Gel of LafutidineFormulation and Evaluation of Floating in Situ Gel of Lafutidine
Formulation and Evaluation of Floating in Situ Gel of Lafutidine
 
Formulation and Evaluation of Floating Matrix Tablets of an Antipsychotic Drug
Formulation and Evaluation of Floating Matrix Tablets of an Antipsychotic DrugFormulation and Evaluation of Floating Matrix Tablets of an Antipsychotic Drug
Formulation and Evaluation of Floating Matrix Tablets of an Antipsychotic Drug
 
Formulation and Evaluation of Gabapentin Mucoadhesive Gastro Retentive Tablets.
Formulation and Evaluation of Gabapentin Mucoadhesive Gastro Retentive Tablets.Formulation and Evaluation of Gabapentin Mucoadhesive Gastro Retentive Tablets.
Formulation and Evaluation of Gabapentin Mucoadhesive Gastro Retentive Tablets.
 
Aarti grdds ppt
Aarti grdds pptAarti grdds ppt
Aarti grdds ppt
 
01_IJPBA_1770_19.pdf
01_IJPBA_1770_19.pdf01_IJPBA_1770_19.pdf
01_IJPBA_1770_19.pdf
 
DEVELOPMENT AND OPTIMIZATION OF CIPROFLOXACIN HCL HOLLOW MICROSPHERES (MICRO ...
DEVELOPMENT AND OPTIMIZATION OF CIPROFLOXACIN HCL HOLLOW MICROSPHERES (MICRO ...DEVELOPMENT AND OPTIMIZATION OF CIPROFLOXACIN HCL HOLLOW MICROSPHERES (MICRO ...
DEVELOPMENT AND OPTIMIZATION OF CIPROFLOXACIN HCL HOLLOW MICROSPHERES (MICRO ...
 
Approaches Of Gastro-Retentive Drug Delivery System or GRDDS
Approaches Of Gastro-Retentive Drug Delivery System or GRDDSApproaches Of Gastro-Retentive Drug Delivery System or GRDDS
Approaches Of Gastro-Retentive Drug Delivery System or GRDDS
 
B
BB
B
 
Formulation Design, Optimization and Evaluation of Carvedilol Phosphate Gastr...
Formulation Design, Optimization and Evaluation of Carvedilol Phosphate Gastr...Formulation Design, Optimization and Evaluation of Carvedilol Phosphate Gastr...
Formulation Design, Optimization and Evaluation of Carvedilol Phosphate Gastr...
 
ausama PP.PDF
ausama PP.PDFausama PP.PDF
ausama PP.PDF
 
01_IJPBA_1770_19.pdf
01_IJPBA_1770_19.pdf01_IJPBA_1770_19.pdf
01_IJPBA_1770_19.pdf
 
01_IJPBA_1770_19.pdf
01_IJPBA_1770_19.pdf01_IJPBA_1770_19.pdf
01_IJPBA_1770_19.pdf
 

1557 full

  • 1.   Kharia et al: Formulation and Evaluation of Polymeric Nanoparticles of an Antiviral Drug for Gastroretention 1557   International Journal of Pharmaceutical Sciences and Nanotechnology Volume 4 •  Issue 4 • January – March 2012 Research Paper MS ID: IJPSN-8-19-11-KHARIA Formulation and Evaluation of Polymeric Nanoparticles of an Antiviral Drug for Gastroretention 1,2 3 1 A.A. Kharia , A.K. Singhai and R. Verma 1 Oriental College of Pharmacy, Bhopal, Madhya Pradesh, India 2 Uttarakhand Technical University, Dehradun, Uttarakhand, India 3 Lakshmi Narayan College of Pharmacy, Bhopal, Madhya Pradesh, India Received August 19, 2011; accepted January 12, 2012 ABSTRACT The aim of present study was to formulate and evaluate nanoparticles of acyclovir by using different hydrophilic polymers. Acyclovir was selected as a suitable drug for gastroretentive nanoparticles due to its short half life, low bioavailability, high frequency of administration, and narrow absorption window in stomach and upper part of GIT. The nano-precipitation method was used to prepare nanoparticles so as to avoid both chlorinated solvents and surfactants to prevent their toxic effect on the body. Nanoparticles of acyclovir were prepared by using hydrophilic polymers such as bovine serum albumin, chitosan, and gelatin. The prepared formulations were then characterized for particle size, polydispersity index, zeta potential, loading efficiency, encapsulation efficiency and drug-excipient compatibility. The prepared nanoparticulate formulations of acyclovir with different polymers in 1:1 ratio have shown particle size in the range of 250.12-743.07 nm, polydispersity index (PDI) in the range of 0.681-1.0, zeta potential in the range of -14.2 to +33.2 mV, loading efficiency in the range of 8.74-17.54%, and entrapment efficiency in the range of 55.7%-74.2%. Nanoparticulate formulation prepared with chitosan in 1:1 ratio showed satisfactory results i.e. average particle size 312.04 nm, polydispersity index 0.681, zeta potential 33.2 mV, loading efficiency 17.54%, and entrapment efficiency 73.4%. FTIR study concluded that no major interaction occurred between the drug and polymers used in the present study. KEYWORDS: Nanoparticles; gastro-retentive; nano-precipitation, polydispersity index, zeta potential; entrapment efficiency. Introduction The oral route of drug administration is the most convenient and commonly used method of drug delivery due to their considerable therapeutic advantages such as ease of administration, patient compliance, and flexibility in formulation (Garg et al., 2008; Gupta et al., 2007). However, this route has several physiological problems, such as inability to restrain and locate the controlled drug delivery system within the desired region of the gastrointestinal tract due to variable gastric emptying and motility. Furthermore, the relatively brief gastric emptying time in humans, which normally means 2-3 hours through the major absorption zone, i.e., stomach and upper part of the intestine, can result in incomplete drug release from the drug delivery system leading to reduced efficacy of the administered dose (Rouge et al., 1996). These difficulties have prompted researchers to design a drug delivery system which can stay in the stomach for prolonged and predictable period (Deshpande et al., 1996; Hwang et al., 1998). Several attempts are being made to develop a controlled drug delivery system, which can provide therapeutically effective plasma drug concentration for a longer period, thereby reducing the dosing frequency and minimizing fluctuations in plasma drug concentration at steady-state by delivering the drug in a controlled and reproducible manner (Sood et al., 2003). Different methodologies have been reported in the literature to increase the gastric retention of drugs, like intra-gastric floating systems, hydro dynamically balanced systems, extendable or expandable, microporous compartment system, microballons, bio/muco-adhesive systems, high-density systems, and super porous biodegradable hydro gel systems (Singh et al., 2000). After oral administration, such a dosage form would be retained in the stomach for several hours and would release the drug there in a controlled and prolonged manner, so that the drug could be supplied continuously to its absorption sites in the upper gastrointestinal tract (Streubel et al., 2006). Prolonged gastric retention improves bioavailability, reduces drug waste, and improves solubility of drugs that are less soluble in a high pH environment. It is also suitable for local drug delivery to the stomach and proximal small intestine (Rao et al., 2005). Gastroretention helps to provide better availability of new products with suitable therapeutic activity and substantial benefits for patients. ABBREVIATIONS: Bovine serum albumin (BSA); molar (M); ultraviolet (UV); Fourier transform infra red (FTIR); polydispersity index (PDI). 1557
  • 2. 1558 Acyclovir, a cyclic analogue of the natural nucleoside 2-deoxyguanosine, is clinically used in the treatment of herpes simplex, varicella zoster, cytomegalovirus, and Epstein Barr virus infections (Brien et al., 1989). Absorption of orally administered acyclovir is slow, variable, and incomplete, with a bioavailability of 15%30% (Shao et al., 1994) and the elimination half-life of acyclovir is approximately 3 hours. It has narrow absorption window and is primarily absorbed from stomach and upper part of the small intestine (Groning et al., 1996); there is a need to develop an effective formulation with enhanced gastric residence time. Attia et al., prepared niosomes of acyclovir by using cholesterol, span 60, and dicetyl phosphate to improve its poor and variable bioavailability. The niosomal dispersion when compared to the free solution has shown more than 2-fold increase in drug bioavailability and increase in the mean residence time of acyclovir reflecting sustained release characteristics (Attia et al., 2007). Palmberger et al., developed a novel oral delivery system for the acyclovir, utilizing thiolated chitosan as excipient, which is capable of inhibiting P-glycoprotein (P-gp). Three chitosan-4-thiobutylamidine (Chito–TBA) conjugates with increasing molecular mass (Chito-9.4 kDa-TBA, Chito-150 kDa-TBA, and Chito-600 kDa-TBA) were synthesized and permeation studies on rat intestinal mucosa and Caco-2 monolayers were performed. They found that Chito-150 kDa–TBA/GSH might be an appropriate sustained release drug delivery system for acyclovir, which is able to enhance acyclovir transport due to efflux pump inhibition (Palmberger et al., 2008). Kharia et al., optimized the floating drug delivery systems of acyclovir using psyllium husk and hydroxypropylmethylcellulose K4M as the polymers by 3² full factorial design to improve the oral bioavailability of acyclovir. The optimized formulations followed Higuchi’s kinetics while the drug release mechanism was found to be anomalous type, controlled by diffusion through the swollen matrix (Kharia et al., 2010). Stulzer et al., prepared microparticles containing acyclovir and chitosan cross-linked with tripolyphosphate using the spray-drying technique. The results obtained indicated that the polymer/acyclovir ratio influenced the final properties of the microparticles, with higher ratios giving the best encapsulation efficiency, dissolution profiles, and stability (Stulzer et al., 2009). The aim of the present study was to formulate gastroretentive nanoparticles of acyclovir to deliver the drug at a controlled rate to its absorption site so that its oral bioavailability can be enhanced. Mucoadhesive polymers, such as bovine serum albumin, chitosan, and gelatin, were selected to prepare gastroretentive nanoparticles as they intensify the contact between dosage form and the site of absorption, thereby reducing the luminal diffusion pathway of the drug (bioadhesion) and lead to significant improvements in oral drug delivery (Lueben et al., 1994, Park and Robinson, 1984). These mucoadhesive polymeric nanoparticles in the stomach will offer various advantages such as (i) Longer residence time of the dosage form on mucosal tissues in Int J Pharm Sci Nanotech Vol 4; Issue 4 • January−March 2012 the stomach. This will improve absorption of the drug and increase the drug bioavailability. (ii) Higher drug concentration at the site of adhesion absorption, which will create a driving force for the paracellular passive uptake. (iii) Immediate absorption from the bioadhesive drug delivery system without previous dilution and possible degradation in the luminal fluids (Hejazi and Amiji et al., 2003) Materials and Method Acyclovir, bovine serum albumin (BSA), and gelatin were obtained as a gift sample from Modern Laboratories (Indore, India); chitosan was obtained as a gift sample from Indian Sea Foods (Cochin, India). All other chemicals and reagents were of laboratory grade and were used as procured. Preparation of Nanoparticles Nanoparticles were prepared according to the nanoprecipitation method with slight modification (Elshafeey et al., 2010). Briefly, 200 mg of polymer (bovine serum albumin, chitosan, and gelatin) was dissolved in 25 ml of acetone separately. The acyclovir 100 mg was dissolved in 2 ml of dimethyl sulfoxide. Both solutions were mixed and then 50 ml of water was added and stirred for a half hour. Acetone was eliminated by evaporation under reduced pressure using rotary flash evaporator and the final volume of the suspension was adjusted to 10 ml. Then this suspension was centrifuged at 15000 rpm at 4oC for half an hour. The supernatant was discarded and precipitate was washed 3 times with distilled water. The nanoparticles thus obtained were dried overnight in oven at 60ºC and stored in a desiccator. The prepared formulations were characterized for loading efficiency, entrapment efficiency, particle size, particle size distribution, polydispersity index, zeta potential and drug excipient compatibility studies. Characterization of Acyclovir Loaded Nanoparticles Loading Efficiency Drug content in the preparation was determined by extracting the drug from the nanoparticles with 0.1 M hydrochloric acid. In this method, the nanoparticles (50 mg) were stirred in 50 ml of 0.1 M hydrochloric acid until dissolved; it was filtered through a Millipore filter and the drug content was determined, after suitable dilution, at 254 nm by UV spectrophotometry. The loading efficiency (L) of the nanoparticles was calculated according to Equation 1 L (%) = (Qn /Wn) × 100 …..(1) Where Wn is the weight of the nanoparticles and Qn is the amount of drug present in the nanoparticles (Patel and Patel, 2007).
  • 3. 1559   Kharia et al: Formulation and Evaluation of Polymeric Nanoparticles of an Antiviral Drug for Gastroretention Drug-loading and entrapment efficiency Entrapment Efficiency For determination of drug entrapment, the amount of drug present in the clear supernatant after centrifugation was determined (w) by UV spectrophotometer at 254 nm. A standard calibration curve of drug was plotted for this purpose. The amount of drug in supernatant was then subtracted from the total amount of drug added during the preparation (W). Effectively, (W-w) will give the amount of drug entrapped in the particles (Bellare et al., 2005). Then percentage entrapment of a drug was calculated according to Equation 2 % Drug Entrapment = (W-w/W) × 100 …..(2) Particle Size, Particle Size Distribution, and Zeta Potential The particle size and particle size distribution of the formulation was determined by photo correlation spectroscopy with a zeta master (Malvern Instruments, UK) equipped with the Malvern PCS software. Every sample was diluted with distilled water. The surface charge (Zeta potential) was determined by measuring the electrophoretic mobility of the nanoparticles using a Malvern zeta sizer (Malvern Instruments, UK). Samples were prepared by diluting with distilled water (Pignatello et al., 2006). Polydispersity Index Polydispersity index is a parameter to define the particle size distribution of nanoparticles obtained from photon correlation spectroscopic analysis. It is a dimensionless number extrapolated from the autocorrelation function and ranges from a value of 0.01 for mono dispersed particles and up to values of 0.5-0.7. Samples with very broad size distribution have polydispersity index values > 0.7 (Nidhin et al., 2008). Drug-Excipient Compatibility Studies The drug excipient compatibility studies was performed by using FT-IR spectrophotometer (Perkin Elmer). The FT-IR spectra of drug, polymers, and formulations were analyzed separately and then correlated for incompatibility. Results and Discussion The method of nanoprecipitation was used so as to avoid both chlorinated solvents and surfactants to prevent their toxic effect on the body. All the determinations were done in triplicate. Although drug loading expresses the percent weight of active ingredient encapsulated to the weight of nanoparticles, entrapment efficiency is the ratio of the experimentally determined percentage of drug content compared with actual, or theoretical mass, of drug used for the preparation of the nanoparticles. The loading efficiency depends on the polymer-drug combination and the method used. Hydrophobic polymers encapsulate larger amounts of hydro phobic drugs, whereas hydrophilic polymers entrap greater amounts of more hydrophilic drugs. Several formulation parameters, such as emulsifier type, weight ratio of polymer to drug, and organic to aqueous phase ratio, will influence the extent of drug loading. The effect of polymer on drug loading efficiency and entrapment efficiency are given in Table 1 and shown in Figure 1. The values were in the range of 8.74%-17.54% and 55.7%-74.2%, respectively. Loading efficiency was low for gelatin and albumin nanoparticles (8.74% and 11.43% respectively) while high for chitosan nanoparticles (17.54%). It was found that the entrapment efficiency were high for the formulations containing chitosan and gelatin (73.4% and 74.2% respectively) while low for the formulation containing bovine serum albumin (55.7%). Loading efficiency may be increased by increasing polymer ratio, so that sufficient quantity of polymer will be available to entrap the drug present in the solution, while less entrapment efficiency may be due to hydrophilic nature of acyclovir. Particle Size Distribution and Polydispersity Index The particle size and particle size distribution are critical factors in the performance of nanoparticles, as batches with wide particle size distribution show significant variations in drug loading, drug release, bioavailability, and efficacy. Particle size and particle size distribution can be determined using light scattering techniques and by scanning or transmission electron microscopy. Formulation of nanoparticles with a narrow size distribution will be a challenge if emulsion cannot be produced with a narrow droplet size distribution. As nanoparticles are internalized into cells by endocytosis, an increase in particle size will decrease uptake and potentially, affect bioavailability of the drug. The extent of endocytosis is dependent on the type of the target cell. TABLE 1 Drug-loading and entrapment efficiency. Sl. No. Formulation Code Polymer Drug:Polymer ratio Formulation Code Loading Efficiency* ± SD % Entrapment Efficiency* ± SD % 1. 2. 3. NP 1 NP 2 NP 3 Bovine Serum Albumin Chitosan Gelatin 1:1 1:1 1:1 NP 1 NP 2 NP 3 11.43±2 17.54±3 8.74±3 55.7±6.3 73.4±3.5 74.2±4.9 * = Average of three determinations
  • 4. 1560 Int J Pharm Sci Nanotech Vol 4; Issue 4 • January−March 2012 Fig. 1. Effect of polymer on loading and entrapment efficiency. The results of prepared nanoparticulate formulations of acyclovir with different polymers are given in Table 2 and shown in Figure 2. The formulations had very high polydispersity index (PDI) in the range of 0.681-1.0. From the particle size distribution data, it is evident that in case of BSA nanoparticles, mean particle diameter was 250.12 nm and major portion of the particles were in the range of 200-400 nm, for chitosan nanoparticles mean particle diameter was 312.04 nm; and major portion of the particles were in range of 200-525 nm. In case of gelatin nanoparticles mean particle diameter was 743.07 nm and most of the particles were in the range of 480-1200 nm. However, in all the formulations contained a minority population of nanoparticles in much smaller range. For BSA, about 10.1% of the particles were in the range 15-30 nm, for chitosan about 7.1% of the particles were in the range 48-90 nm and for gelatin 14.1% of the particles were in the range 70-160 nm. These minority populations are responsible for larger over all polydispersity indices of the formulations. We are currently exploring the process variables affecting the relative amounts of different populations with an objective to increase the yield of the particles in the smaller range to get much smaller nanoparticles, which have greater degree of monodispersity. Such nanoparticles can be easily separated from the larger sized population by simple methods like filtration. From the above data it is clear that nanoparticles prepared by using chitosan and BSA exhibited reduction in mean nanoparticulate diameter and narrower granulometric distribution. But the nanoparticles prepared using gelatin as a polymer resulted in nanoparticulate population of large particles. The higher particle size and polydispersity index may be because of absence of emulsifier as the use of emulsifier decreases the surface tension between organic phase acetone and aqueous phase and leads to the formation of smaller solvent droplets, which in turn causes decrease in particle size. It also stabilizes newly generated surfaces and prevents aggregation of the particles as reported by previous researchers (Schubert and Muller, 2003). Therefore results which were obtained in this study may be improved by using increased drug:polymer ratio, using different formulation strategy such as desolvation (for gelatin and albumin) or counter ion induced aggregation (for chitosan and sodium alginate), employing cross linking agent followed by neutralizing residual cross linking agent with cysteine and high speed stirring. Zeta Potential The measurement of the zeta potential allows predictions about the storage stability of colloidal dispersions. In general, particle aggregation is less likely to occur for charged particles (i.e. high zeta potential) due to electric repulsion. Generally, Zeta potential values above 30 mV (positive or negative values) lead to more stable nanocapsule suspensions because repulsion between the particles prevented their aggregation. A decrease in zeta potential, i.e. electrostatic repulsion, was considered as the cause for the aggregation process (Dalengon et al., 1998). The charge on the surface of the nanospheres will influence their distribution in the body and the extent of uptake into the cells. Because cell membranes are negatively charged, there is greater electrostatic affinity for positively charged nanoparticles. Therefore, the surface of cationic or neutral nanoparticles may be modified to confer a positive charge to enhance efficacy. The zeta potential values which were in the range of –14.2 - +21.7 mV, indicates that the colloidal suspension may not be stable and may lead to aggregation. Zeta potential values can be altered by modifying the major components such as surfactants, polymer, and surface composition of the nanoparticles, the presence or the absence of adsorbed compounds, composition of the dispersing phase, mainly the ionic strength, and the pH (Barratt et al., 1999).
  • 5. 1561   Kharia et al: Formulation and Evaluation of Polymeric Nanoparticles of an Antiviral Drug for Gastroretention during the process of formulation polymer has not reacted with the drug to give rise to reactant products. So it is only physical mixture and there is no interaction between them which is in favor to proceed for formulation. Drug-Excipient Compatibility Studies The results of drug-excipient compatibility studies are shown in Table 3. From the IR data it is clear that functionalities of drug have remained unchanged, including intensities of the peak. This suggests that TABLE 2 Drug polymer ratio, mean particle size, particle size distribution, poly dispersity index (PDI) and zeta potential. Sl. No. Formulation Code Polymer Mean Particle Size (nm) ± SD Size Distribution PDI ± SD Zeta Potential (mV) ± SD 1. NP 1 Bovine Serum Albumin 250.12±18 10.1% (15-30 nm) 89.9 % (200-400 nm) 1.0±0.12 21.7±1.4 2. NP 2 Chitosan 312.04±32 7.8% (48-90 nm) 92.2% (200-525 nm) 0.681±0.15 33.2±2.1 3. NP 3 Gelatin 743.07±45 14.2% (70-160 nm) 85.8% (480-1200 nm) 0.7770±0.14 -14.2±1.3 * = Average of three determination Fig. 2. Effect of polymer on mean particle size. TABLE 3 Major peaks observed in the FT-IR spectrum. S. No. 1. 2. 3. 4. 5. 6. 7. 8. Bands -OH N-H bend, assy. C-C ring str. Aromatic amines, C-N str. Assy. C-O-C str. Sym C-O-C str. N-H wagging Mono-substitution in ring Drug NP 1 cm-1 cm-1 NP 2 NP 3 cm-1 3447.8 and 3337.9 1632.5 cm-1 1537 and 1483.7 cm-1 1305.9 and 1392.1 cm-1 3442.9 1626.4 cm-1 1479.2 and 1531.1 cm-1 1387.1 cm-1 3434.5 and 3324.5 1638.8 cm-1 1456.9 cm-1 1390.4 and 1302.5 cm-1 3441.1 cm-1 1635.4 cm-1 1532.1 and 1481.1 cm-1 1303.6 cm-1 1219 cm-1 1013 cm-1 900.5 and 864.2 cm-1 777.1,753.7 and 681.2 cm-1 1216.8 cm-1 1011.9 cm-1 900.1 cm-1 767.6 and 672 cm-1 1219.2 cm-1 1080.3 cm-1 897.3 cm-1 771.5 and 675.5 cm-1 1216.5 cm-1 1011.2 cm-1 899.5 cm-1 769.8 and 670.9 cm-1 NP1 = Bovine Serum Albumin Nanoparticles, NP2 = Chitosan Nanoparticles, NP3 = Gelatin Nanoparticles Conclusion Among different nanoparticulate formulations prepared by nanoprecipitation method formulation NP 2, with chitosan in 1:1 drug: polymer ratio, showed satisfactory results; i.e. mean particle size of 312.04 nm (majority of the particles were in the range of 200-525 nm), polydispersity index of 0.681, zeta potential of 33.2 and loading efficiency of 17.54%, and entrapment efficiency of 73.4%. FTIR study concluded that no major interaction occurred between the drug and polymers used in the present study.
  • 6. 1562 Acknowledgements The authors are thankful to Modern Laboratories (Indore, India) and Indian Sea Foods (Cochin, India) for providing gift samples and Director, Oriental College of Pharmacy, Bhopal, India for their support and cooperation in carrying out the research work. The authors are very thankful to SAIL, RGPV, Bhopal, for their valuable contribution in the determination of particle size, PDI and zeta potential of the formulations. References Attia IA, El-Giawy SA, Fouda MA and Donia AM (2007). Influence of a niosomal formulation on the oral bioavailability of acyclovir in rabbits. AAPS Pharm SciTech 8; 106: E1-E27. Barratt G (1999). Characterization of colloidal drug carrier systems with zeta potential measurements. Pharma Tech Eur 11: 25–32. Bellare J, Banerjee R and Das S (2005). Aspirin loaded albumin nanoparticles by coacervation: Implications in drug delivery. Trends Bio Artif Organs 18: 203-211. Dalengon F, Amjaud Y, Lafforgue C, Derouin F, Fessi H (1998). Atovaquone and rifabutine-loaded nanocapsules: Formulation studies. Int J Pharm 153: 127-130. Deshpande AA, Rhodes TN and Shah H (1996). Controlled release drug delivery systems for prolonged gastric residence an overview. Drug dev Ind pharm 22: 531-539. Elshafeey AH, Kamel AO and Awad GAS (2010). Ammonium methacrylate unit’s polymer content and their effect on acyclovir colloidal nanoparticles properties and bioavailability in human volunteers. Colloids and Surf B: Biointerfaces 75: 398-404. Garg R and Gupta GD (2008). Progress in controlled gastroretentive delivery systems. Trop J Pharm Res 7: 1055-1066. Groning R, Berntgen M and Geogarakis M (1996). Acyclovir serum concentrations following peroral administration of magnetic depot tablets and the influence of extra corporal magnet to control gastrointestinal transit. Eur J Pharm Biopharm 46: 28591. Gupta SN and Aggarwal N (2007). A gastro-retentive floating delivery system for 5-Fluorouracil. Asian Journal of Pharmaceutical Sciences 2: 143-149. Hejazi R and Amiji M (2003). Chitosan-based gastrointestinal delivery systems. J Control Release 89: 151-165. Hwang SJ, Park H and Park K (1998). Gastric retentive drug delivery systems. Crit Rev Ther Drug 15: 243-248. Kharia AA, Hiremath SN, Singhai AK, Omray LK, Jain SK (2010). Design and optimization of floating drug delivery system of acyclovir. Indian J Pharm Sci 72: 599-606. Lueben HL, Lehr CM, Rentel CO, Noach ABJ, Boer AGD, Verhoef JC and Junginger HE (1994). Bioadhesive polymers for the peroral delivery of peptide drugs. J Control Release 29: 329-338 Int J Pharm Sci Nanotech Vol 4; Issue 4 • January−March 2012 Nidhin M, Indumathy R, Sreeram KJ and Balachandran U (2008). Synthesis of iron oxide nanoparticles of narrow size distribution on polysaccharide templates. Bull Mater Sci 31: 93-96. O’Brien J and Campoli D (1989). Acyclovir: an updated review of its antiviral activity, pharmacokinetics properties and therapeutic efficacy. Drugs 37: 233-309. Palmberger TF, Hpmbach J and Schnurch AB (2008). Thiolated chitosan: Development and in vitro evaluation of an oral delivery system for acyclovir. Int J Pharm 348: 54-60. Park K and Robinson JR (1984). Bioadhesive polymers as platforms for oral-controlled drug delivery: method to study bioadhesion. Int J Pharm 19: 107-127. Patel JK and Patel MM (2007). Stomach Specific Anti-Helicobacter Pylori Therapy: Preparation and Evaluation of Amoxicillin – Loaded Chitosan Muco adhesive Microspheres. Curr Drug Deliv 4: 41-50. Pignatello R, Ricupero N, Bucolo, Maugeri F, Maltese A and Puglisi G (2006). Preparation and characterization of eudragit retard nanasuspensions for the ocular delivery of cloricromene. AAPS Pharm SciTech 24; 7: E27. Rao PB, Kottan NA and Snehith VS (2005). Development of gastroretentive drug delivery system of cephalexin by using factorial design. ARS Pharmaceutica 50: 8-24. Rouge N, Buri P and Doelke E (1996). Drug absorption sites in the gastrointestinal tract and dosage forms for site specific delivery. Int J Pharm 136:117-139. Schubert MA and Muller GCC. Solvent injection as a new approach for manufacturing lipid nanoparticles-evaluation of the method and process parameters. Eur J Pharm Biopharm 2003;55:125131 Shao Z and Mitra AK (1994). Bile salts-fatty acid mixed micelles as nasal absorption promotors: III. Effect on nasal transport and enzymatic degradation of acyclovir prodrug. Pharm Res 11: 24350. Singh BN and Kim KH (2000). Floating drug delivery systems: an approach to oral controlled drug delivery via gastric retention. J Control Release 63: 235-239. Sood A and Panchagnula R (2003). Design of controlled release delivery systems using modified pharmacokinetic approach: a case study for drugs having a short elimination half life and a narrow therapeutic index. Int J Pharm 261: 27-41. Streubel A, Siepmann J and Bodmeier R (2006). Gastroretentive drug delivery system. Expert Opin Drug Deliv 3: 217-233. Stulzer HK, Tagliari MP, Parize AL, Silva MAS and Laranjeira MCM (2009). Evaluation of cross-linked chitosan microparticles containing acyclovir obtained by spray-drying. Mater Sci Eng C 29: 387-392. Address correspondence to: Ankit Anand Kharia, Oriental College of Pharmacy Raisen Road, Thakral Nagar, BHOPAL (M.P.) 462021. Mob: 09685360536; Email: ankitanandkharia@yahoo.co.in