Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Drug Development and Industrial Pharmacy, 2013; 39(4): 540–547

© 2013 Informa Healthcare USA, Inc.


ISSN 0363-9045 print/ISSN 1520-5762 online
DOI: 10.3109/03639045.2012.666978

Research article

Chitosan based mucoadhesive nanoparticles of ketoconazole


for bioavailability enhancement: formulation, optimization,
in vitro and ex vivo evaluation
Drug Development and Industrial Pharmacy Downloaded from informahealthcare.com by McMaster University on 03/11/13

Jigar Modi, Garima Joshi, and Krutika Sawant

TIFAC Centre of Relevance and Excellence in NDDS, Centre of PG Studies and Research, Pharmacy Department,
The M. S. University of Baroda, Fatehgunj, Vadodara, Gujarat, India

Abstract
Purpose: The conventional dosage form of Ketoconazole (KZ) shows poor absorption due to rapid gastric emptying.
Chitosan based mucoadhesive nanoparticles (NPs) of KZ were developed to efficiently release drug at its absorption
window i.e. stomach and the site of action i.e. esophagus.
Method: The NPs were prepared by ionic gelation method. Concentration of polymer, cross-linking agent and ratio of
drug/polymer as well as polymer/cross linking agent were optimized.
For personal use only.

Results: NPs had 69.16 ± 5.91% mucin binding efficiency, particle size of 382.6 ± 2.384 nm, ζ potential of +48.1 mv
and entrapment efficiency of 59.84 ± 1.088%. DSC thermogram indicated absence of any drug polymer interaction.
The drug release was by controlled, non-fickian diffusion mechanism. Ex vivo diffusion studies were performed by
emptying the stomach contents after 2 h to simulate in vivo gastric emptying. The results showed that drug diffusion
from the solution across stomach mucosa stopped after emptying whereas that from the NPs continued upto 5 h.
Hence we could conclude that the NPs must have adhered to the stomach mucosa and thereby would have been
retained at this absorption site even after gastric emptying.
Conclusion: The orally delivered KZ loaded mucoadhesive NPs can be used as an efficient carrier for delivering drug at
its absorption window i.e. the stomach and the site of action i.e. esophagus even after gastric emptying.
Keywords:  Chitosan, ionic gelation, ketoconazole, Ex vivo, nanoparticles

Introduction
(HIV/AIDS), acid suppression, esophageal dysmotility,
Candidiasis has emerged as a significant medical prob- gastric surgery and antibiotic use3.
lem associated with indiscriminate long-term use of Systemically active therapy is needed for effective
antibiotics, immunosuppressive and cytotoxic therapies, treatment of Candida esophagitis, for which Azole based
immune-defects and acquired immunodeficiency syn- antifungal agents are drugs of choice. Ketoconazole
drome (AIDS)1. The GI tract is the major site of dissemi- (KZ) is a broad spectrum antifungal agent useful in the
nated candidiasis2. The esophagus is the most common treatment of candidal esophagitis. However, it requires
site of colonization of fungi. Candida esophagitis is most an acidic pH for absorption and its absorption is depen-
common type of infective esophagitis found to occur in dent on gastric acidity and gastric emptying time, lead-
patients with predisposing factors including malignancy, ing to variable bioavailability. Though it is an orally
corticosteroid use, diabetes mellitus, human immuno- active drug, its clinical use is sometimes limited due to
deficiency virus/acquired immunodeficiency syndrome its erratic absorption. Often, the major drawback of the

Address for Correspondence:  Krutika Sawant, TIFAC Centre of Relevance and Excellence in NDDS, Centre of PG Studies and Research,
Pharmacy Department, G. H. Patel Pharmacy Building, Donor’s Plaza, The M. S. University of Baroda, Fatehgunj, Vadodara-390002, Gujarat,
India. Tel: +91 265 2434187. Fax: +91 265 2418927. E-mail: dr_krutikasawant@rediffmail.com
(Received 08 September 2011; revised 20 December 2011; accepted 13 February 2012)

540
Orally delivered chitosan nanoparticles of ketoconazole  541

Abbreviations TPP: Tripolyphosphate


PDE: Percentage drug entrapped
NPs: Nanoparticles TEM: Transmission electron microscopy
CS: Chitosan DSC: Differential scanning calorimetry
KZ: Ketoconazole

conventional dosage form is that due to rapid gastric Thus it was hypothesized that TPP cross-linked
emptying, the absorption of the drug is poor (Cmax after Chitosan nanoparticles would improve absorption and
a single oral dose of 200 mg is reached within 2 h and is bioavailability of KZ which requires acidic pH for absorp-
3.5 mcg/mL)4. tion and whose conventional dosage form often shows
Drug Development and Industrial Pharmacy Downloaded from informahealthcare.com by McMaster University on 03/11/13

Polymeric nanoparticles (NPs) have been extensively poor absorption of drug due to rapid gastric emptying.
researched as drug delivery carriers5. NPs are suitable Also, use of TPP in chitosan NPs may synergize the anti-
carriers to deliver drugs which are unstable in the biolog- fungal activity of Ketoconazole.
ical fluids and cannot readily traffic across the mucosal Therefore, the present study was aimed to develop and
barrier6. Polymeric NPs from biodegradable polymers optimize orally delivered mucoadhesive chitosan nano-
are effective drug carriers because they are expected to particles of KZ which will efficiently release drug at its
be absorbed intact after oral administration7. Oral NPs absorption window i.e. stomach and near its site of action
are drug delivery systems of choice due to enhanced (esophagus). The optimized formulation was character-
bioavailability, mucoadhesion and controlled release of ized for particle size, % drug entrapment, ζ potential,
drugs in GIT8,9. Mucoadhesion has become the area of Differential scanning calorimetry (DSC) and TEM. The
interest of researchers for its potential to optimize local- formulations were evaluated for mucin binding capac-
ized drug delivery, by retaining a dosage form at the ity, drug diffusion study through dialysis membrane and
absorption window in the GIT. However, the significant through rat stomach and obtained data were fitted to
problem with large mucoadhesive solid dosage forms Korsmeyer Peppas model to understand the mechanism
For personal use only.

like tablets is the poor adhesion to the mucosal surfaces of drug release.
due to their large size and the vigorous movement of
the GIT10. Hence, NPs are considered to be much better
Materials and methods
alternative to overcome such problem.
NPs may be used for oral administration of gut- Materials
labile drugs or those with low aqueous solubility11. The KZ was obtained as gift sample from Gufic Bioscience Pvt.
NPs are expected to adhere to the gastric mucosa and Ltd, Mumbai, India. Low molecular weight CS (molecu-
remain in the gastrointestinal tract, while protecting the lar weight less than 60 kDa) (deacetylation value >85%),
entrapped drug from enzymatic degradation, until the viscosity 30–50 cps was acquired from Ample Effect Sdn
release of the loaded drug or their absorption as intact Bhd, Malaysia, and was used without any modification
particles12. Chitosan (CS) has the potential of serving and purification. Sodium Tripolyphosphate was pur-
as an absorption enhancer across intestinal epithelia chased from Loba Chemie, Mumbai, India. Pig Mucin
due to its mucoadhesive and permeability enhancing was acquired from Sigma-Aldrich, St Louis, USA. Glacial
property13. It has biocompatibility, biodegradability, acetic acid, Mannitol and all other chemicals used were
high charge density and non toxicity14. CS NPs loaded of analytical grade.
with insulin showed increased oral bioavailability of
the peptide and prolonged reduction in blood glucose Preparation of NPs
levels by protecting the labile drug from enzymatic deg- The ionic gelation method was used for preparation of
radation in GI tract15. CS is soluble at acidic pH but has KZ loaded CS NPs20. In brief, CS (30 mg) was dissolved
limited solubility at neutral as well as basic pH 16. Hence, in 1% acetic acid solution and hydrated overnight with
it can be used as absorption enhancer for drugs which stirring. KZ (6 mg) was dissolved in the CS solution with
are more soluble in the acidic environment of stomach. stirring. Aqueous solution of TPP (7.5 mL, 1 mg/mL) was
Ionic gelation technique has attracted considerable then added drop wise to the CS solution (10 mL, 3 mg/
attention as this process is organic solvent free, con- mL) under constant stirring on magnetic stirrer (Remi
venient and controllable17. Sodium tripolyphosphate Equipment Pvt. Ltd., Mumbai, India), which led to the
(TPP) is the most extensively used ion cross-linking formation of dispersion of NPs. The nanodispersion
agent due to its non-toxic and multivalent properties18. was then centrifuged at 25,000 rpm for 30 min (3K 30,
A recent study by Palmeira-de Oliveira et  al reported Sigma Laboratory Centrifuge, Osterode, Germany) and
that TPP had an inhibitory species dependent and the pellet of the NPs was redispersed in water. The NPs
concentration dependent cytotoxicity against several were lyophilized for 24 h (Lyophillizer, Heto Dry Winner,
Candida spp. Strains19. Allerod, Denmark) using mannitol as cryoprotectant

© 2013 Informa Healthcare USA, Inc.


542  J. Modi et al.
[NPs (1 parts): cryoprotectant (3 parts)] to get free flow- Germany). The supernatant was diluted with appropri-
ing powder of NPs. ate amount of 0.1 N HCl and analyzed for the amount
The various process parameters were optimized sys- of unentrapped drug by UV-Visible spectrophotometer
tematically. Only one parameter was changed at a time (UV-1601, Shimadzu, Kyoto, Japan) at 270 nm. Similarly,
in each set of experiments. The parameters studied the pellet was dissolved in 0.1 N HCl and analyzed for the
included concentration of polymer, concentration of entrapped drug content by UV-Visible spectrophotom-
cross-linking agent and ratios of drug/polymer as well as eter at 270 nm21.
polymer/cross-linking agent. The effect of these variables The percentage drug entrapped (PDE) was calculated
on particle size and entrapment efficiency was evaluated according to following formula:
to obtain optimized nanoparticulate formulation with
minimum possible size and maximum possible entrap- PDE ( % ) = ( TD-FD/TD ) ×100
ment. All batches were prepared in triplicate.  Where TD is total amount of drug added and FD is
amount of free drug in supernatant.
Drug Development and Industrial Pharmacy Downloaded from informahealthcare.com by McMaster University on 03/11/13

Characterization of NPs
Particle size Measurement of mucoadhesive force
Size of the CS NPs was measured by dynamic light The mucoadhesive properties of drug loaded CS NPs
scattering using Malvern Zetasizer Nano ZS (Malvern were evaluated by measuring the amount (% weight) of
Instruments, Malvern, Worcestershire, UK). A suitable Pig Mucin (Sigma-Aldrich, St Louis, MO, USA) bound to
amount of NPs was dispersed in distilled water creat- the NPs as per the method reported by Yin et al. (2006)22.
ing a total concentration of 1% w/v. Each sample was In brief, 2 mL of mucin solution (100 µg/mL) was mixed
measured in triplicate and particle size was expressed as with 2 mL NPs suspension, incubated at 37 °C for 30 min
mean diameter ± standard deviation. and centrifuged at 25000 rpm for 30 min (3K 30, Sigma
Laboratory centrifuge, Osterode, Germany). The super-
ζ potential natant was separated and the free mucin content was
ζ potential distribution was measured using a Zetasizer determined by UV spectrophotometry at 251 nm. The %
(Nano ZS, Malvern Instruments, Malvern, Worcestershire, mucin binding efficiency of the NPs was calculated using
UK). Each sample was suitably diluted with filtered dis-
For personal use only.

the following formula:


tilled water creating a concentration of 1% w/v and placed
in a disposable ζ cell. The electrophoretic mobility (µm/ % mucin binding efficiency = (Co − Cs ) /Co  ×100
sec) was converted to ζ potential by in-built software using
Helmholtz-Smoluchowski equation. Three measurements  Where,
of each sample were used to derive average ζ potential. Co = Initial concentration of mucin
CS = Concentration of free mucin in the supernatant
Transmission electron microscopy
The morphology of drug loaded NPs was ascertained by In vitro diffusion study
Transmission electron microscopy (TEM). The studies In vitro diffusion study was carried out in order to deter-
were performed using Transmission electron microscope mine the kinetics and mechanism of drug release from
(Phillips Technai-20, Hillsboro, OR) at 50000 X. NPs were the CS NPs. The study was carried out in phosphate buf-
dispersed in distilled water and one drop of the reconsti- fer saline (PBS) (pH 7.4) containing 1% sodium lauryl
tuted NPs was incubated on carbon coated copper grid. sulphate (SLS) for a period of 5 h, as it is reported that the
The copper grid was fixed into sample holder and placed mucus layer is continuously renewed and replaced by a
in the vacuum chamber of the transmission electron fresh layer; the turnover time of the gastric mucus is of
microscope and observed under low vacuum. 4–5 h from production to digestion23.
The suspension of NPs equivalent to 2 mg drug was
Differential scanning calorimetry placed in a previously activated dialysis bag (cut off
The physical state of KZ loaded in NPs was investigated molecular weight: 12000 Daltons, Himedia, India) and
using DSC. The analysis was carried out using a DSC was sealed at both the ends. The bag was then placed in
(DSC 60, Shimadzu, Kyoto, Japan) at a heating rate of a beaker containing 20 mL of diffusion medium which
20°C per min in the range of 30°C to 300°C under inert was maintained at 37 ± 2°C and stirred continuously at
nitrogen atmosphere at a flow rate of 40 mL/min. DSC 100 rpm24. At regular intervals, samples were withdrawn
thermograms were recorded for CS, KZ and KZ loaded from the receptor phase and replaced with the same
CS NPs. quantity of fresh medium to maintain sink conditions25.
All samples were analyzed spectroscopically at 270 nm to
Entrapment efficiency determine the amount of drug released. All the experi-
The entrapment efficiency was determined after sepa- ments were repeated thrice and the average values were
rating the NPs from the aqueous medium (containing recorded. The same process was carried out for the plain
non-entrapped KZ) by centrifugation at 25000 rpm for drug by taking 2 mL of KZ solution (1 mg/mL in 0.1 N
30 min (3K 30, Sigma Laboratory Centrifuge, Osterode, HCl) as control.

 Drug Development and Industrial Pharmacy


Orally delivered chitosan nanoparticles of ketoconazole  543
The kinetic analysis of the release data was done using was filled into the stomach. The esophageal connection
Korsmeyer and Peppas or power law equation (Peppas was then tied and the stomach was mounted in the
et al. 1987)26 organ tube containing 20 mL PBS (pH 7.4) with 1% SLS.
Temperature was maintained at 37 ± 0.5°C using heat-
M t / M ∞ = K ⋅t n ing coil. Oxygen was supplied to the tissue through aer-
ator. Samples were withdrawn from the organ tube at
Log ( M t / M ∞ ) = Log K + n log t predetermined intervals and replaced with same quan-
tity of fresh media. After 2 h, the stomach was removed
 Where, time t from the organ bath, opened and the contents were
Mt/M∞ is the fractional drug release, K is a kinetic con- emptied (to simulate in vivo gastric emptying, which is
stant incorporating structural and geometrical character- 2 h). The stomach was then filled with 0.1 N HCl, tied
istic of the drug/polymer system, n is diffusion exponent and again mounted in the organ bath and the diffu-
that characterizes the mechanism of drug release, t is the sion was continued till 5 h. All samples were analyzed
Drug Development and Industrial Pharmacy Downloaded from informahealthcare.com by McMaster University on 03/11/13

time. spectroscopically at 270 nm to determine the amount


of drug diffused. The same procedure was repeated for
Ex vivo diffusion study through rat stomach plain drug solution27.
This study was conducted to obtain idea about drug dif-
fusion across stomach mucosa under conditions simu-
lating in vivo gastric emptying. The ex vivo study was Results and discussion
carried out for a period of 5 h. However, as the in vivo Preparation and optimization of NPs
gastric emptying time is 2 h, the stomach contents were The basic principle behind the formation of CS NPs
emptied after 2 h and diffusion studies were continued in Ionic Gelation method28 is based on electrostatic
upto 5 h. The studies were carried out according to the interaction between CS and TPP. The NPs form due to
guidelines of Committee for the Purpose of Control intermolecular linkages between amino groups of CS
and Supervision of Experiments on Animals, Ministry and phosphate groups of TPP29. CS and TPP concentra-
of Social Justice and Empowerment, Government tions play an important role in the formation of NPs.
of India, New Delhi, India and were approved by the Furthermore, CS reacts with TPP in fixed ratios only and
For personal use only.

Institutional Animal Ethics Committee of Pharmacy it should be within the range of 4:1–6:1 to obtain NPs30.
Department. Male albino rats weighing 200–250 g were Hence, these formulation parameters were optimized
fasted overnight, sacrificed by spinal dislocation and on the basis of particle size and PDE and the results are
the stomach was isolated immediately. The stomach shown in Tables 1–3. Various concentrations of CS (2,
contents were emptied and it was cleaned with normal 2.5, 3 and 4 mg/mL) were used. At a fixed weight ratio28
saline. Then the lower ends of the stomach (duodenal of CS/TPP, ionic gelation between CS and TPP is affected
connection) were tied with thread and from the esoph- by concentration of CS, thus affecting the particle size
ageal connection, 2 mL nanoparticulate formulation of the NPs31. With increasing CS concentration, particle

Table 1.  Optimization of CS concentration.


CS/TPP weight ratio CS/Drug ratio CS conc. (mg/mL) TPP conc. (mg/mL) Particle size* (nm) PDE* (%)
4:1 4:1 2.0 1.0 177.2 ± 3.164 20.22 ± 1.950
4:1 4:1 2.5 1.0 284.7 ± 1.352 28.64 ± 1.339
4:1 4:1 3.0 1.0 364.0 ± 4.218 42.31 ± 1.136
*Data represents mean ± S.D. (n = 3).

Table 2.  Optimization of TPP concentration.


CS/TPP weight ratio CS/Drug ratio CS conc. (mg/mL) TPP conc. (mg/mL) Particle size* (nm) PDE* (%)
4:1 4:1 3.0 0.5 316.4 ± 2.155 30.52 ± 1.451
4:1 4:1 3.0 0.75 348.2 ± 2.463 34.291 ± 0.311
4:1 4:1 3.0 1.0 364.0 ± 4.218 42.311 ± 0.136
*Data represents mean ± S.D. (n = 3).

Table 3.  Optimization of CS/TPP weight ratio and CS/Drug ratio.


CS/TPP weight ratio CS/Drug ratio CS conc. (mg/mL) TPP conc. (mg/mL) Particle Size* (nm) PDE* (%)
4:1 4:1 3.0 1.0 364.0 ± 4.218 42.31 ± 1.136
4:1 5:1 3.0 1.0 382.6 ± 2.384 59.84 ± 1.088
5:1 4:1 3.0 1.0 312.52 ± 0.116 34.13 ± 1.225
5:1 5:1 3.0 1.0 358.4 ± 3.912 44.59 ± 1.455
*Data represents mean ± S.D. (n = 3).

© 2013 Informa Healthcare USA, Inc.


544  J. Modi et al.
size was found to increase which could be attributed to
increase in viscosity of CS solution. Very high concentra-
tion of CS (4 mg/mL) resulted in formation of micropar-
ticles, because of very high viscosity of the CS solution.
The entrapment efficiency also increased with increase
in CS concentration. This was due to increase in amount
of polymer available for both drug entrapment and for
gelation.
When various concentrations of TPP from 0.5 to
1.25 mg/mL were investigated, it was observed that as
concentration of TPP increased, particle size increased.
However, at 1.25  mg/mL, there was formation of
microparticles. Simultaneously, entrapment increased
Drug Development and Industrial Pharmacy Downloaded from informahealthcare.com by McMaster University on 03/11/13

with increase in TPP concentration. This could be due


to increase in number of TPP ions available to cross-link
amino groups on CS chains20,29, thereby improving gela-
tion and drug incorporation. Figure 1. Transmission electronic micrographs of optimized
The size and the size distribution of the NPs depend ketoconazole loaded chitosan nanoparticles.
upon the weight ratio between CS and TPP32. For this
reason, the effect of CS/TPP weight ratio on the size char- particulate formulation in vitro, but also affects the
acteristics of the NPs was studied in order to find the opti- interactions with biomolecules in vivo. The high posi-
mum ratio that yields NPs of moderate size and narrow tive ζ value is advantageous as it indicates high muco-
size distribution. However, it was observed that though adhesive interaction with mucin which contains sialic
there was a reduction in size due to more compact parti- acid and sulphonic acid residues that are negatively
cle structure, entrapment efficiency also decreased upon charged35. Thus, the ζ potential of the NPs was suitable
increasing the CS/TPP weight ratio from 4:1 to 5:1, prob- for providing physical stability as well as for mucoad-
ably due to inefficient crosslinking having taken place at hesion which would improve in vivo drug permeation
For personal use only.

higher CS contents. There are less TPP ions available to across the mucosal membrane.
cross-link the higher content of Chitosan. Particle size
and entrapment efficiency was also dependent on CS/ Differential scanning calorimetry
drug ratio. As CS/drug ratio increased, both, particle size Differential scanning calorimetry (DSC) is an impor-
and entrapment increased due to greater amount of CS tant technique for investigation of thermal properties
available for incorporating the drug. CS/drug ratio of 5:1 of a formulation and provides information about the
provided maximum drug entrapment (59.84 ± 1.088%) physicochemical state of drug in the system. There is no
with moderate particle size (382.6 ± 2.384 nm). Hence, detectable endotherm if the drug is present in a molec-
the batch containing CS/TPP weight ratio of 4:1 and CS/ ular dispersion or solid solution state in the polymeric
Drug ratio of 5:1 was considered as optimized batch (at NPs36. The DSC curves of CS, KZ and drug loaded CS NPs
CS conc. 3 mg/mL and TPP conc. 1 mg/mL). were obtained. It was seen that the DSC thermograms
of drug loaded CS NPs showed a broad endothermic
Characterization of NPs peak at 102.81°C which was due to the glass transition
Particle size, morphology and ζ potential temperature of CS. However, the melting peak of drug
Particle size of the optimized batch was found to was absent in the thermogram of NPs indicating that KZ
be 382.6 ± 2.384 nm. Drug loaded NPs must have an was incorporated in amorphous form in the CS matrix
appropriate particle size along with ζ potential. The in the NPs.
size characteristics have been found to affect the bio-
logical performance of CS NPs33. TEM image showed Measurement of mucoadhesive force
that NPs were spherical in shape without aggregation The mucoadhesive properties of CS NPs were evalu-
(Figure 1). The ζ potential value is important as it can ated by measuring the pig mucin binding efficiency on
influence particle stability as well as mucoadhesivity. the NPs which was found to be 69.16 ± 5.91%. From this
The more positive and negative values of ζ potential value, we expect that the NPs will adhere to the mucosal
tend to stabilize particle suspension to a greater extent membrane in vivo and thereby improve absorption and
as the electrostatic repulsion between particles of like bioavailability of KZ. The high value of binding efficiency
charges prevents the aggregation of particles34. The can be attributed to highly positive ζ potential, the
ζ potential of the drug loaded NPs was found to be bioadhesive nature of CS and the nano size of the NPs.
+48.1 mv indicating physical stability of the system. The Studies on isolated porcine gastric mucosa preparations
high positive ζ was attributed to the positively charged have shown that hydrated CS has a positive charge and
CS (due to protonation of its amino groups in acetic can adhere to negatively charged mucus gel layers. This
acid). ζ potential not only governs the stability of the ability could result in CS containing formulations being

 Drug Development and Industrial Pharmacy


Orally delivered chitosan nanoparticles of ketoconazole  545
retained in the stomach. Adhesion would be expected to the plot of log Mt/M∞ versus log t for NPs was found to be
be particularly marked under the acidic conditions in the 0.9791, indicating a good model fit26. The nanoparticulate
stomach, where cationic CS would be highly charged37. formulation exhibited a non-fickian diffusion mecha-
Therefore, it implies that the formulation will be retained nism as the value of release exponent ‘n’ was found to
at the absorption window in the stomach and KZ will be be more than 0.5 (n = 0.9042). The release of drug from
better absorbed from there. NPs was initially by diffusion followed by erosion. As CS
is a hydrophilic, swellable polymer, it can be assumed
In vitro drug release study that the drug release was controlled by combination of
The efficiency of conventional dosage form of KZ is polymer swelling, diffusion through the hydrated matrix
limited due to short gastric residence time. As CS has and erosion.
mucoadhesive potential, the drug can be better retained
at absorption window for prolonged period. Comparison Ex-vivo diffusion study
of release profile of plain drug solution and NPs formu- The ex vivo studies are very important to understand
Drug Development and Industrial Pharmacy Downloaded from informahealthcare.com by McMaster University on 03/11/13

lation showed that sustained release of KZ was obtained the real drug release characteristics through natural
from the NPs (Figure 2) due to the cross-linking between organs and tissues38. Hence, we designed ex vivo diffu-
amino groups of CS and the phosphate ions of TPP29. sion studies aimed at simulating actual in vivo condi-
Sustained drug release from mucoadhesive NPs is impor- tions wherein any ingested substance is subjected to
tant as it would allow for a prolonged residence time at average gastric emptying after 2 h.Thus, the stomach
the absorption window. Thus, 99.63 ± 1.23% of drug was contents were emptied after 2 h of starting the ex vivo
released from the drug solution while 80.36 ± 2.15% of diffusion studies. The data showed that 53.22 ± 2.19% of
drug was released from NPs by the end of 5 h. Although drug was released from plain drug solution at the end
the drug release from plain drug solution was more than of 2 h (Figure 3). However, when the stomach contents
90%, it should be noted that the plain drug formula- were emptied after 2 h, the drug release from plain drug
tion would leave the absorption window (i.e. stomach) solution stopped, indicating its complete removal from
at around 2 h because of gastric emptying and hence the stomach due to emptying. On the other hand, drug
the effective absorption would be much less than the release continued from the NPs formulation even after
observed data. On the other hand, the nanoparticulate emptying the contents after 2 h, with 78.37 ± 2.24% drug
For personal use only.

formulation will remain adhered to the gastric mucosa release at the end of 5 h. It can thus be concluded from
and will release the drug in controlled manner for a pro- this study that the NP formulation will remain adhered
longed period at the absorption window. Hence, we can to the absorption site i.e. stomach for a prolonged
hypothesize that the entire drug from NPs will be avail- period even after gastric emptying of the stomach
able for absorption at the absorption window and there- contents, and thereby improve the absorption and bio-
fore its bioavailability will be enhanced. Moreover, the availability of KZ. The data obtained from ex vivo drug
presence of the drug near its site of action (esophagus) release studies was fitted to Korsmeyer–Peppas model.
for a prolonged time is expected to improve its therapeu- The regression coefficient of the plot of log Mt/M∞ ver-
tic efficacy sus log t for NPs was found to be 0.9901, indicating a
Theoretically, the drug release from NPs may take good model fit. As expected due to the swelling and
place by several mechanisms including surface ero- erodible nature of CS, the nanoparticulate formulation
sion, disintegration, diffusion and desorption38. Hence exhibited a non-fickian diffusion mechanism as the
to understand the exact mechanism of release, the data value of release exponent n was found to be more than
obtained from in vitro drug release studies were fitted to 0.5 (n = 0.8401).
Korsmeyer–Peppas model. The regression coefficient of

Figure 3. Release profile of KZ from plain drug solution and


Figure 2. Release profile of KZ from plain drug solution and nanoparticulate formulation through Rat stomach, in PBS (pH
nanoparticulate formulation through dialysis bag in PBS (7.4). 7.4) for a period of 5 h.

© 2013 Informa Healthcare USA, Inc.


546  J. Modi et al.

Conclusion  8. Chen H, Langer R. (1998). Oral particulate delivery: status and
future trends. Adv Drug Deliv Rev, 34:339–350.
CS NPs of KZ were prepared by ionic gelation method   9. Kawashima Y. (2001). Nanoparticulate systems for improved drug
using sodium tripolyphosphate as cross-linking agent. delivery. Adv Drug Deliv Rev, 47:1–2.
10. Andrews GP, Laverty TP, Jones DS. (2009). Mucoadhesive polymeric
The concentration of CS, concentration of TPP and CS platforms for controlled drug delivery. Eur J Pharm Biopharm,
/TPP weight ratio were optimized to obtain minimum 71:505–518.
possible size and maximum drug entrapment. The NPs 11. Woolfson AD, Elliott GR, Gilligan CA, Passmore CM. (1999).
showed good entrapment, moderate particle size and Design of an intravaginal ring for the controlled delivery of
high positive ζ potential. DSC studies confirmed the 17 beta-estradiol as its 3-acetate ester. J Control Release,
61:319–328.
absence of any interaction between drug and polymer 12. Arbós P, Campanero MA, Arangoa MA, Renedo MJ, Irache JM.
and indicated the incorporation of drug in polymer (2003). Influence of the surface characteristics of PVM/MA
matrix in an amorphous form. Mucoadhesion studies nanoparticles on their bioadhesive properties. J Control Release,
revealed good mucin binding efficiency and hence 89:19–30.
Drug Development and Industrial Pharmacy Downloaded from informahealthcare.com by McMaster University on 03/11/13

good mucoadhesion capacity of NPs. In vitro and ex 13. Janes KA, Calvo P, Alonso MJ. (2001). Polysaccharide colloidal
particles as delivery systems for macromolecules. Adv Drug Deliv
vivo drug release studies through rat stomach for 5 h Rev, 47:83–97.
showed that the drug release was sustained and fol- 14. Arias JL, López-Viota M, Gallardo V, Adolfina Ruiz M. (2010).
lowed non-fickian transport of drug from NPs. Results Chitosan nanoparticles as a new delivery system for the
of ex vivo diffusion studies designed to simulate actual chemotherapy agent tegafur. Drug Dev Ind Pharm, 36:744–750.
in vivo conditions wherein any ingested substance is 15. Damge C, Michel C, Aprahamian M, Couvreur P, Devissaguet JP.
(1990). Nanocapsules as carriers for oral peptide delivery. J Control
subjected to average gastric emptying showed that the Release, 13:233–239.
drug release from plain drug solution stopped after 2 h, 16. Peng X, Zhang L, Kennedy JF. (2006). Release behaviour
indicating its complete removal from the stomach due of microspheres from cross-linked N-methylated chitosan
to emptying. On the other hand, drug release continued encapsulated ofloxacin. Carbohydr polym, 65:288–295.
from the NPs formulation even after emptying the con- 17. Kim S, Lee J. (2011). Folate-targeted drug-delivery systems prepared
by nano-comminution. Drug Dev Ind Pharm, 37:131–138.
tents. It can thus be concluded from this study that the 18. Lin A, Chen J, Liu Y, Deng S, Wu Z, Huang Y et  al. (2009).
NP formulation will remain adhered to the absorption Preparation and evaluation of N-caproyl chitosan nanoparticles
site i.e. stomach and the site of action i.e. esophagus for surface modified with glycyrrhizin for hepatocyte targeting. Drug
For personal use only.

a prolonged period even after gastric emptying of the Dev Ind Pharm, 35:1348–1355.
stomach contents, and thereby improve the absorption, 19. Palmeira-de-Oliveira R, Palmeira-de-Oliveira A, Gaspar C,
Silvestre S, Martinez-de-Oliveira J, Amaral MH, Breitenfeld L.
bioavailability and therapeutic efficacy of KZ. These (2011). Sodium Tripolyphosphate: An excipient with intrinsic in
findings suggest the suitability of orally delivered KZ vitro anti-Candida activity, Int J Pharm 421:130– 134.
loaded mucoadhesive NPs as efficient carrier system 20. Calvo P, Remunan-Lopez C,Vila-Jato JL, Alonso MJ. (1997). Novel
for delivering drug at absorption window even after hydrophilic chitosan-polyethylene oxide Nanoparticles as protein
gastric emptying. carriers. J App Polymer Sci, 63:125–132.
21. Agnihotri SA, Aminabhavi TM. (2007). Chitosan nanoparticles
for prolonged delivery of timolol maleate. Drug Dev Ind Pharm,
33:1254–1262.
Declaration of interest 22. Yin Y, Chen D, Qiao M, Lu Z, Hu H. (2006). Preparation and
The authors report no conflict of interests. evaluation of lectin-conjugated PLGA nanoparticles for oral
delivery of thymopentin. J Control Release, 116:337–345.
23. Washington N, Washington C, Wilson, CG. (2003). Biological
References barriers to drug absorption in: Physiological Pharmaceutics, 2nd
edition, London: Taylor and Francis, 94–95.
 1. Khan Z, Gyanchandani A (1998). Candidiasis − A Review. Proc 24. Magenheim B, Levy MY, Benita S. (1993). A new in-vitro technique
Indian Natl Sci Acad B, 64:1–34. for the evaluation of drug release profile from colloidal carriers-
 2. Walsh TJ, Merz WG. (1986). Pathologic features in the human ultrafilteration at low pressure. Int J Pharm, 94:115–123.
alimentary tract associated with invasiveness of Candida tropicalis. 25. Joseph NM, Sharma PK. (2007). Cross-linked nanoparticles of
Am J Clin Pathol, 85:498–502. cytarabine: encapsulation, storage and in-vitro release. Afr J Pharm
 3. Mimidis K, Papadopoulos V, Margaritis V, Thomopoulos K, Pharmacol, 1:11–13.
Gatopoulou A, Nikolopoulou V et  al. (2005). Predisposing 26. Peppas NA, Ritger PL. (1987). A simple equation for description
factors and clinical symptoms in HIV-negative patients with of solute release II. Fickian and anomalous release from swellable
Candida oesophagitis: are they always present? Int J Clin Pract, devices. J Control Release, 5:37–42.
59:210–213. 27. Yin Y, Chen D, Qiao M, Wei X, Hu H. (2007). Lectin-conjugated
 4. Martindale (2002). The Complete Drug Reference. (33rd ed). PLGA nanoparticles loaded with thymopentin: ex vivo
London: Pharmaceutical press, 389–390. bioadhesion and in vivo biodistribution. J Control Release,
 5. Gref R, Minamitake Y, Peracchia MT, Trubetskoy V, Torchilin 123:27–38.
V, Langer R. (1994). Biodegradable long-circulating polymeric 28. Malhotra M, Kulamarva A, Sebak S, Paul A, Bhathena J, Mirzaei
nanospheres. Science, 263:1600–1603. M et  al. (2009). Ultrafine chitosan nanoparticles as an efficient
 6. Alonso MJ. (2004). Nanomedicines for overcoming biological nucleic acid delivery system targeting neuronal cells. Drug Dev
barriers. Biomed Pharmacother, 58:168–172. Ind Pharm, 35:719–726.
  7. Florence AT, Hillery AM, Hussain N, Jani PU. (1995). Nanoparticles 29. Ajun W, Yan S, Li G, Huilli L. (2009). Preparation of aspirin and
as carriers for oral peptide absorption: studies on particle uptake probucol in combination loaded chitosan nanoparticles and in
and fate. J Control Release, 36:39–46. vitro release study. Carbohydr Polym, 75:566–574.

 Drug Development and Industrial Pharmacy


Orally delivered chitosan nanoparticles of ketoconazole  547
30. Zhang H, Oh M, Allen C, Kumacheva E. (2004). Monodisperse chitosan nanoparticulate formulations: a potential oral delivery system for
nanoparticles for mucosal drug delivery. Biomacromolecules, hormone therapy. Pharm Res, 23:184–195.
5:2461–2468. 35. Capra RH, Baruzzi AM, Quinzani LM, Strumia MC. (2007).
31. Wu Y, Yang W, Wang C, Hu J, Fu S. (2005). Chitosan nanoparticles Rheological, dielectric and diffusion analysis of mucin/carbopol
as a novel delivery system for ammonium glycyrrhizinate. Int J matrices used in amperometric biosensors. Sensors and Actuator
Pharm, 295:235–245. B:Chemical, 124:466–476.
32. Papadimitriou S, Bikiaris D, Avgoustakis K, Karavas E, Georgarakis 36. Dubernet C. (1995). Thermoanalysis of microspheres. Thermochim
M. (2008). Chitosan nanoparticles loaded with dorzolamide and Acta, 248:259–269.
pramipexole. Carbohydr Polym, 73:44–54. 37. Lehr CM, Bouwstra JA, Schacht EH, Junginger HE. (1992). In vitro
33. PanY, Li Y, Zhao H, Zheng J, Xu H, Wei G, Hao JS, Fu C. (2002) evaluation of mucoadhesive properties of chitosan and some
Bioadhesive polysaccharide in protein delivery system: Chitosan other natural polymers. Int J Pharm, 78:43–48.
nanoparticles improve the absorption of insulin in vivo. Int J 38. Bernkop-Schnürch A, Weithaler A, Albrecht K, Greimel A. (2006).
Pharm, 249:139–147. Thiomers: preparation and in vitro evaluation of a mucoadhesive
34. Hariharan S, Bhardwaj V, Bala I, Sitterberg J, Bakowsky U, nanoparticulate drug delivery system. Int J Pharm, 317:
Ravi Kumar MN. (2006). Design of estradiol loaded PLGA 76–81.
Drug Development and Industrial Pharmacy Downloaded from informahealthcare.com by McMaster University on 03/11/13
For personal use only.

© 2013 Informa Healthcare USA, Inc.

You might also like